Cancer-Fighting Agents

Evidence-informed compounds and repurposed drugs.

Note: These research summaries are informational, not endorsements. Evidence levels vary from lab studies to early human trials. Effects may not translate to any or all humans and some compounds can cause harm. Always consult your doctor before use. Ratings are not purely objective and were determined based on lab results and strength of available evidence, not guaranteed clinical benefit. We make no claim that anything here is curative of any condition.

Key

🔥 Potency1–10

Antitumor effect at realistic exposure.

Example8.0 / 10
🛡 Selectivity1–10

Tumor targeting vs. healthy-tissue hit (therapeutic window).

Example7.0 / 10
🎯 Translatability1–10

Practical to achieve effective exposure (formulation, PK, safety, access, DDIs).

Example9.0 / 10
🧪 Confidence0–5

Strength/consistency of evidence (human data weighted highest).

4 / 5
🎯 Endpoint

Primary outcome/biomarker this agent aims to move (e.g., OS, PFS, ORR; or mechanistic: GSH↓, NADPH↓, FAO↓, xCT↓, Hedgehog↓).

📌 Anchor

Best-supported form/route/dose of the agent (or closest clinical analogue) used to benchmark scores and cite first. Example: Lentinan (IV), Auranofin 3 mg PO qd, Metformin IR 500–1000 mg PO.

6-Shogaol (Ginger)

PreclinicalROSApoptosisNF-κBEMT

Simple Summary: This ginger compound pushes cancer cells into overwhelming stress: it floods them with ROS, jams their protein-folding machinery, and flips on death switches—while turning down pathways they use to invade and spread. It also hits stem-like tumor cells that seed relapse. Evidence is mostly lab/animal so far, but it’s a strong adjunct candidate in preclinical work.

Strength of Evidence: ⭐⭐ Preliminary — Robust cell/animal data across tumor types; human cancer trials are limited/absent, so clinical efficacy is unproven.

Mechanisms: 6-Shogaol, the dehydrated form of 6-gingerol, kills cancer cells by triggering mitochondrial/ER-stress apoptosis driven by reactive oxygen species (ROS) and the PERK–eIF2α–ATF4–CHOP axis (up to DR5 activation). It suppresses NF-κB/IKK and STAT3 signaling, lowering MMP-2/MMP-9 and EMT drivers (Snail/Slug/vimentin), which reduces invasion and metastasis. It also targets cancer stem-like cells (e.g., CD44+CD24−/low/ALDH1+ spheres) and can induce paraptosis-like vacuolization. In models, 6-shogaol enhances sensitivity of drug-resistant cells (e.g., gefitinib-resistant ovarian) via ER-stress–mediated apoptosis.

Apoptosislab
🔥 Potency7.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.5 / 10
🎯 Translatability5.5 / 10
Endpoint: Annexin V @24h
Anchor: Staurosporine 1 μM = 10
ER-stress→DR5; strongest in TNBC/ovarian lines
ROSlab
🔥 Potency7.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5.5 / 10
Endpoint: DCF-DA ROS fluorescence
Anchor: H2O2 100 μM = 10
Mito/ER ROS at ≤10 μM
NF-κBlab
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability5.5 / 10
Endpoint: p65 nuclear translocation / MMP-9
Anchor: BAY 11-7082 5 μM = 10
IKKβ block; consistent across lines
EMTlab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5 / 10
Endpoint: Vimentin/SNAIL; invasion
Anchor: SB431542 10 μM = 10
Colorectal/OV lines; migration↓

Note: Human evidence: Very limited; no clinical trials proving anti-cancer benefits in humans. Ginger extracts (containing 6-shogaol) have been studied for nausea in cancer patients, but not for tumor effects. Safety: Generally safe in food/tea amounts (e.g., from fresh ginger). High doses or extracts may cause GI upset (heartburn, diarrhea), mouth irritation, or bleeding risk (due to anti-platelet effects—avoid with anticoagulants like warfarin). Not recommended in pregnancy or with gallstones. Bioavailability is low; nanotechnology may improve in future. Adjunct role: Potential to enhance chemo (e.g., in resistant cells), but discuss with oncologist to avoid interactions. Do not self-administer high doses without supervision.

References:

Acemannan

PreclinicalImmune ↑Macrophage Activator

Simple Summary: Acemannan is a compound found in aloe vera that boosts your immune system’s cancer-fighting activity. It helps the body recognize tumors and slows their growth. One human study even showed better outcomes when acemannan was used with chemo.

Strength of Evidence: ⭐⭐⭐ Moderate — Multiple preclinical studies plus one human trial.

Mechanisms: Acemannan stimulates macrophages, dendritic cells, and T-cells to release IL-1, IL-6, TNF-α, and IFN-γ, enhancing the immune system’s ability to recognize and attack tumors. It also promotes antigen presentation and slows tumor proliferation, migration, and stemness in vitro. In combination with chemotherapy, it may enhance tumor regression and improve survival.

Immunehuman
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity8.0 / 10
🎯 Translatability7.5 / 10
Endpoint: NK cytotoxicity / IFN-γ
Anchor: IL-2 (clinical) = 10
Oral aloe polysaccharide; human signals with chemo
Macrophage Activatorlab
🔥 Potency6.3 / 10
🧪 Confidence3 / 5
🛡 Selectivity7.5 / 10
🎯 Translatability7.5 / 10
Endpoint: TNF-α/IL-1β release; MHC-II
Anchor: GM-CSF (ref) = 10
Dectin/TLR bias; non-toxic range

Note: All but one of these studies were performed in mice or canines. The human trial (PMID 19368145) was larger and concluded: “The percentage of both objective tumor regressions and disease control was significantly higher in patients concomitantly treated with Aloe than with chemotherapy alone, as well as the percent of 3-year survival patients.” In that trial, patients received 10 ml of aloe extract three times daily alongside chemotherapy.

References:

Active Hexose Correlated Compound (AHCC)

Early humanNKT-cellCytokines ↑ApoptosisImmune modulationTumor growth ↓

Simple Summary: AHCC, a shiitake mushroom extract, supercharges your immune system's NK and T-cells to better spot and destroy cancer cells. It also nudges tumors toward self-destruction and teams up with chemo to make it more effective while easing side effects like nausea or low blood counts. Human studies show promise for liver, pancreatic, and ovarian cancers, especially as an add-on to standard treatments.

Strength of Evidence: ⭐⭐⭐ Moderate — Preliminary RCTs/cohorts in HCC (improved survival), pancreatic/ovarian (reduced toxicities, immune boost); strong mechanistic/preclinical data; larger oncology trials needed.

Mechanisms: AHCC is a proprietary alpha-glucan-rich extract from cultured shiitake (Lentinula edodes) mycelia, with low-molecular-weight polysaccharides (primarily alpha-1,4-glucans with alpha-1,3 branches, ~5 kDa). It modulates immunity by priming Toll-like receptors (TLR2/TLR4) on intestinal epithelium and immune cells, enhancing dendritic cell maturation, NK cell cytotoxicity, and T-cell (CD4+/CD8+) proliferation. This leads to upregulated cytokine production (IL-12, IFN-γ, TNF-α) and nitric oxide regulation. Anticancer effects include induction of tumor cell apoptosis (Bax↑/Bcl-2↓, caspase-3/7 activation), overcoming multidrug resistance via downregulation of Heat Shock Factor 1 (HSF1) and HSP27, and synergy with chemotherapy (e.g., gemcitabine, 5-FU) by improving drug efficacy and reducing toxicities. It also exhibits anti-inflammatory and antioxidant properties, protecting against oxidative stress and chemo-induced adverse events. Pharmacokinetically, AHCC is well-absorbed orally with good bioavailability, peaking in 1-2h and exerting systemic immune effects.

NK activationhuman
🔥 Potency7.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.5 / 10
🎯 Translatability8 / 10
Endpoint: NK cytotoxicity assay
Anchor: IL-2 100 IU/mL = 10
Via IL-12/IFN-γ; consistent in cancer patients
T-cell responsehuman
🔥 Potency7.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7.5 / 10
Endpoint: CD4+/CD8+ proliferation
Anchor: Anti-CD3/CD28 beads = 10
Enhanced during chemo
Cytokineshuman
🔥 Potency6.8 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability8 / 10
Endpoint: IL-12/IFN-γ ELISA
Anchor: LPS 1 μg/mL = 10
Pro-inflammatory shift for anti-tumor
Apoptosislab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: Annexin V / caspase-3
Anchor: Staurosporine 1 μM = 10
Bax/Bcl-2 modulation in tumor cells
Immune modulationhuman
🔥 Potency7.2 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8.5 / 10
Endpoint: TLR2/4 expression / DC maturation
Anchor: BCG vaccine = 10
Intestinal priming → systemic effects
Tumor growthhuman
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability7 / 10
Endpoint: Tumor volume / recurrence rate
Anchor: 5-FU 10 μM = 10
Adjunct in HCC cohorts

Note: Ideal as an immune-boosting adjunct during chemo; take on empty stomach for best absorption. Monitor for CYP2D6 induction with certain drugs. Not a standalone cure—pair with conventional care under provider guidance. Limited long-term oncology data; ongoing trials may clarify broader use.

References:

Agaricus Blazei

Early humanβ-GlucanNK

Simple Summary: This medicinal mushroom helps wake up your immune system. It supercharges the cells that fight cancer—especially NK cells—and can help shrink tumors in early studies. Some clinical trials have shown improvements in immune markers for cancer patients who take it.

Strength of Evidence: ⭐⭐⭐ Moderate — Multiple small human studies and strong mechanistic animal data.

Mechanisms: Agaricus blazei is rich in β-glucans that activate Dectin-1 and TLR-2 receptors on immune cells, leading to the production of IL-12 and IFN-γ. This stimulates natural killer (NK) cells, macrophages, and cytotoxic T-cells. In vitro and in vivo studies show it inhibits tumor growth, induces apoptosis, and may reverse immunosuppression in cancer patients.

β-Glucanhuman
🔥 Potency6.8 / 10
🧪 Confidence4 / 5
🛡 Selectivity8.5 / 10
🎯 Translatability8 / 10
Endpoint: Dectin-1/TLR-2 activation; IL-12
Anchor: Lentinan (IV) = 10
Oral extracts raise β-glucan activity; good safety
NKhuman
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity8.5 / 10
🎯 Translatability8 / 10
Endpoint: NK killing % / IFN-γ
Anchor: IL-2 (clinical) = 10
Small trials: NK function↑; QoL and immune markers↑

Note: Human evidence includes small trials showing immune marker improvements (e.g., NK activity ↑) and QoL benefits in cancer patients, but no large RCTs for tumor outcomes. Preclinical data strong for antitumor effects. Safety good up to 5.4 g/day in phase I trials; monitor for GI effects.

References:

Aged Garlic Extract (AGE)

Early humanApoptosisAngiogenesisNKDNA Damage ↓Inflammation ↓

Simple Summary: This standardized garlic extract pushes cancer cells toward programmed death, starves tumors by cutting off new blood vessels, and supports immune attack (NK cells). A small clinical trial suggests it can slow growth of colon polyps, but direct tumor-shrinkage evidence in people is still limited.

Strength of Evidence: ⭐⭐⭐ Moderate — Small randomized/controlled human studies (adenomas; immune activation) plus strong mechanistic/animal data. Direct anti-cancer efficacy in patients is not yet established.

Mechanisms: AGE is enriched in stable, water-soluble organosulfurs—especially S-allylcysteine (SAC) and S-allyl-mercaptocysteine (SAMC)—that trigger mitochondrial apoptosis (Δψm loss, Bax↑/Bcl-2↓ → caspase-9/-3 activation), arrest the cell cycle, and suppress pro-tumor pathways (NF-κB, COX-2). It inhibits angiogenesis (↓VEGF/VEGFR2; impaired endothelial migration/tube formation) and reduces invasion in colorectal and other models. In humans, AGE increased natural-killer (NK) cell number/activity in advanced digestive-cancer patients and, in a randomized trial, slowed the growth/number of colorectal adenomas (precancerous lesions).

Apoptosishuman
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8 / 10
Endpoint: Annexin V @24h
Anchor: Staurosporine 1 μM = 10
SAMC/SAC mitochondrial apoptosis; oral SAC achievable
Angiogenesishuman
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8 / 10
Endpoint: Endothelial tube formation / VEGF signaling
Anchor: Sunitinib 50–100 nM = 10
VEGF/VEGFR2 and migration↓ in models; adenoma data supports prevention
NKhuman
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity8.5 / 10
🎯 Translatability8.5 / 10
Endpoint: NK cytotoxicity / count
Anchor: IL-2 (clinical) = 10
↑NK number/activity in digestive-cancer patients
DNA Damagelab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability8 / 10
Endpoint: γH2AX / comet assay
Anchor: N-acetylcysteine (DNA damage markers) = 10
Reduces oxidative DNA damage markers; context-dependent with ROS therapies
Inflammationhuman
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability8.5 / 10
Endpoint: NF-κB/COX-2/CRP surrogate
Anchor: BAY 11-7082 5 μM (NF-κB) = 10
NF-κB/COX-2 down; anti-inflammatory signals in humans

Note: Why AGE > raw garlic for cancer: AGE provides consistent, standardized SAC (stable, bioavailable, low-toxicity) with human data for adenoma suppression and immune activation. Raw garlic’s key compound allicin is highly unstable and varies with crushing/cooking, making dose and effects unpredictable.

References:

Albendazole

PreclinicalMicrotubule DisruptionApoptosis Induction

Simple Summary: Albendazole is a common deworming drug now studied for cancer because it interferes with tumor cell division. Lab tests show it shrinks tumors in models of colon, liver, and skin cancers, and a small human trial noted some benefits but warned of blood cell risks.

Strength of Evidence: ⭐⭐ Moderate — Robust preclinical activity; preliminary human signals overshadowed by toxicity.

Mechanisms: Albendazole binds to β-tubulin, disrupting microtubule formation and causing mitotic arrest, which leads to apoptosis and inhibition of glucose uptake in cancer cells. It suppresses NF-κB signaling to reduce inflammation and angiogenesis, and promotes ubiquitin-mediated PD-L1 degradation to enhance immunotherapy. Preclinical models demonstrate anti-proliferative effects in liver, colon, pancreatic, melanoma, and other cancers; human data show potential but highlight hematologic toxicity.

Microtubule Disruptionpreclinical
🔥 Potency7.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.5 / 10
🎯 Translatability7 / 10
Endpoint: Tubulin polymerization / mitotic arrest
Anchor: Paclitaxel (clinical) = 10
β-tubulin binding; glucose uptake inhibition
Apoptosis Inductionlab
🔥 Potency7.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability6.5 / 10
Endpoint: Caspase activation / Bcl-2 ↓
Anchor: Withaferin A (in vitro) = 10
Mitotic stress-induced; NF-κB suppression

Note: Strong preclinical evidence across multiple cancer types; human data limited to small pilot studies (e.g., PMID 11474247: 7 patients with advanced HCC/CRC, some tumor marker stabilization but 3/10 developed severe neutropenia, 1 possibly treatment-related death). Promising for repurposing but requires careful monitoring for myelosuppression.

References:

Alpha-Lipoic Acid

Early humanRedoxHIF-1α ↓AKTNF-κB

Simple Summary: ALA recharges the body’s own antioxidants and turns down cancer growth switches (AKT, NF-κB, HIF-1α). That can reduce blood-vessel growth and glycolysis and help tumor cells self-destruct. Human studies mostly show symptom/metabolic benefits; direct tumor-control evidence is still early.

Strength of Evidence: ⭐⭐⭐ Moderate — Supported by multiple human studies, mechanistic trials, and animal models. Human studies focus mostly on quality of life improvements, while anti-tumor activity remains mostly preclinical.

Mechanisms: Alpha-lipoic acid (ALA) is a mitochondrial coenzyme and redox regulator that restores glutathione, vitamin C, and vitamin E. It inhibits tumor-promoting pathways such as AKT, NF-κB, and HIF-1α, suppressing angiogenesis and glycolysis in cancer cells. ALA has been shown to induce apoptosis, arrest the cell cycle, and reduce oxidative stress in various cancer types including ovarian, lung, and breast cancer models.

Redox↔︎human
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity3.5 / 10
🎯 Translatability9 / 10
Endpoint: GSH/GSSG; antioxidant network restoration
Anchor: N-acetylcysteine (restorative redox) = 10
Restores GSH, Vit C/E; may blunt ROS-based hits—time away from ROS waves
HIF-1αlab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability8.5 / 10
Endpoint: HIF-1α protein / target genes
Anchor: Acriflavine (HIF-1 dimerization) = 10
Suppresses hypoxia signaling → anti-angiogenic/glycolysis effects
AKTlab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability8.5 / 10
Endpoint: p-AKT (S473)
Anchor: LY294002 10 μM = 10
Downstream growth signaling moderation
NF-κBhuman
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability9 / 10
Endpoint: p65 nuclear translocation / cytokines
Anchor: BAY 11-7082 5 μM = 10
Anti-inflammatory signaling in human studies; QoL/metabolic benefits common
References:

Andrographolide (Andrographis paniculata)

Early humanNF-κBSTAT3HIF-1α ↓VEGFMMP-2/9 ↓PI3K/Akt/mTORCOX-2ApoptosisAutophagyROSInvasion/MetastasisAngiogenesis

Simple Summary: Andrographolide turns down master survival switches (NF-κB, STAT3) and hypoxia signaling (HIF-1α/VEGF), pushes cancer cells toward apoptosis, slows the cell cycle, and often blocks PI3K/Akt/mTOR. It also cuts invasion programs like MMP-2/9 and CXCR4. Because oral levels are low and short-lived in people, it’s best viewed as a chemo-sensitizer/adjunct rather than a stand-alone anticancer drug right now.

Strength of Evidence: ⭐⭐ Low to Moderate — Robust mechanistic and preclinical data (NF-κB/STAT3/HIF-1α, PI3K/Akt/mTOR, anti-angiogenesis/invasion); limited human oncology data and constrained oral bioavailability.

Mechanisms: Andrographolide is a diterpenoid lactone that acts as a Michael acceptor. It directly and covalently modifies NF-κB p50 at Cys62 to block NF-κB DNA binding, suppressing transcription of survival, inflammatory, and pro-metastatic genes (e.g., COX-2, MMP-9). It inhibits JAK/STAT3 signaling (sensitizing cells to doxorubicin and other cytotoxics), downregulates HIF-1α (via PI3K/Akt suppression and ubiquitin-mediated degradation) and thereby lowers VEGF and angiogenesis. Across tumor models it induces mitochondrial apoptosis (caspase-3/7 activation, PARP cleavage, Bax↑/Bcl-2↓), triggers G0/G1 or G2/M arrest, and modulates autophagy in a context-dependent way (most often via PI3K/Akt/mTOR inhibition). It also reduces migration/invasion by lowering MMP-2/9, CXCR4, HER2 and related programs. PK in humans shows low oral bioavailability and short half-life; most clinical data are non-oncology, so anticancer efficacy in humans remains unproven.

NF-κBlab
🔥 Potency7.2 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability4.5 / 10
Endpoint: p50/p65 DNA binding
Anchor: BAY 11-7082 5 μM = 10
Covalent p50 Cys62 modification
STAT3lab
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4.5 / 10
Endpoint: p-STAT3 (Y705) / reporter
Anchor: Stattic 5 μM = 10
Chemo-sensitization reported
HIF-1αlab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4.5 / 10
Endpoint: HIF-1α protein/targets
Anchor: Acriflavine 5 μM = 10
Via PI3K/Akt suppression & degradation
VEGFlab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5 / 10
Endpoint: VEGF secretion / tube formation
Anchor: Sunitinib 50–100 nM = 10
Endothelial effects
MMP-2/9lab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5 / 10
Endpoint: Gelatin zymography / invasion
Anchor: Marimastat 1 μM = 10
Migration/invasion ↓
PI3K/Akt/mTORlab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4.5 / 10
Endpoint: p-AKT / p-mTOR
Anchor: LY294002 10 μM = 10
Growth pathway block
COX-2lab
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability5 / 10
Endpoint: COX-2/PGE2
Anchor: Celecoxib 10 μM = 10
Anti-inflammatory
Apoptosislab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability4.5 / 10
Endpoint: Annexin V / caspase-3/7
Anchor: Staurosporine 1 μM = 10
Mitochondrial pathway
Autophagylab
🔥 Potency5.0 / 10
🧪 Confidence2 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability4.5 / 10
Endpoint: LC3-II / flux
Anchor: Rapamycin 100 nM = 10
Context-dependent (inhibit vs induce)
ROSlab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability4.5 / 10
Endpoint: DCF-DA
Anchor: H2O2 100 μM = 10
Pro-oxidant stress in tumors
Invasion/Metastasislab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability5 / 10
Endpoint: Matrigel invasion
Anchor: SB431542 10 μM = 10
EMT-related suppression
Angiogenesislab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability5.5 / 10
Endpoint: Endothelial tube formation
Anchor: Sunitinib 50–100 nM = 10
VEGF-linked

Note: Consider for protocols aiming to suppress NF-κB/STAT3/HIF-1α and PI3K/Akt/mTOR, reduce angiogenesis/invasion, and sensitize to chemo or ROS hits. Watch for context-dependent autophagy effects (can inhibit or, with some analogs/conditions, induce it). Human cancer outcome trials are lacking; oral bioavailability is limited—formulation and timing matter.

References:

Apigenin

Simple Summary: Apigenin slows cancer-cell division, pushes them toward self-destruction, and reduces blood-vessel growth. It dials down PI3K/AKT and NF-κB and can raise ROS inside tumor cells. Most evidence is lab/animal; concentrated extracts are needed for pharmacologic effects beyond diet.

Strength of Evidence: ⭐⭐ Preclinical — Strong cell and animal data; human trials are limited or lacking.

Mechanisms: Apigenin is a flavonoid that exerts anticancer effects by inducing G2/M cell cycle arrest, inhibiting angiogenesis (via VEGF downregulation), and acting as a topoisomerase-II poison. It modulates key signaling pathways such as PI3K/AKT, MAPK, and NF-κB, inhibits cancer cell migration and invasion, promotes apoptosis through mitochondrial disruption, and increases intracellular ROS generation selectively in cancer cells.

PI3Klab
🔥 Potency5.6 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: p-AKT (S473)
Anchor: LY294002 10 μM = 10
Growth signaling moderation
NF-κBlab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: p65 nuclear translocation
Anchor: BAY 11-7082 5 μM = 10
Anti-inflammatory gene suppression
Angiogenesislab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: Tube formation / VEGF
Anchor: Sunitinib 50–100 nM = 10
VEGF pathway down
ROSlab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability6 / 10
Endpoint: DCF-DA
Anchor: H2O2 100 μM = 10
Preferential ROS in tumor cells

Note: Best for protocols targeting PI3K/NF-κB suppression, angiogenesis, or ROS sensitization. Dietary sources provide low exposure; supplements aim for 50-200 mg/day. Limited human oncology data—use as adjunct with monitoring. Biphasic ROS effects: low-dose protective, higher cytotoxic.

References:

Apricot Seed / Amygdalin

Simple Summary: Amygdalin can release cyanide after enzymatic conversion, damaging mitochondria and triggering apoptosis. While it shows tumor cell kill in lab models, human trials have not shown benefit and cyanide toxicity is a real risk. If considered at all, it must be under medical supervision with strict dosing and monitoring—or avoided.

Strength of Evidence: ⭐⭐ Preclinical — Strong mechanistic and animal data, but human trials are sparse or controversial.

Mechanisms: Amygdalin, found in bitter apricot seeds, is converted by β-glucosidase (often elevated in cancer cells) into benzaldehyde and hydrogen cyanide, which disrupt mitochondrial function and induce apoptosis. It downregulates anti-apoptotic Bcl-2, activates caspase-3, increases ROS production, and disrupts cellular metabolism. Some studies also suggest anti-proliferative and anti-angiogenic effects in specific tumor lines.

Mito Toxinlab
🔥 Potency3.5 / 10
🧪 Confidence2 / 5
🛡 Selectivity1.5 / 10
🎯 Translatability2.5 / 10
Endpoint: Δψm loss (JC-1/TMRM)
Anchor: Rotenone 1 μM = 10
Cyanide-mediated ETC inhibition; potency constrained by safety
Bcl-2lab
🔥 Potency4.0 / 10
🧪 Confidence2 / 5
🛡 Selectivity2.0 / 10
🎯 Translatability2.5 / 10
Endpoint: Bcl-2 protein/activity
Anchor: Venetoclax 100 nM = 10
Indirect downregulation vs direct Bcl-2 inhibitor
Caspaselab
🔥 Potency4.2 / 10
🧪 Confidence2 / 5
🛡 Selectivity2.0 / 10
🎯 Translatability2.5 / 10
Endpoint: Caspase-3/7 activity / Annexin V
Anchor: Staurosporine 1 μM = 10
Apoptosis secondary to mitochondrial injury
ROSlab
🔥 Potency4.0 / 10
🧪 Confidence2 / 5
🛡 Selectivity2.0 / 10
🎯 Translatability2.5 / 10
Endpoint: DCF-DA
Anchor: H2O2 100 μM = 10
ROS rise accompanies mitochondrial damage; non-selective

Note: Warning: a 1982 study on stage 4 patients showed no improvement in outcomes, and some patients had cyanide toxicity. Another more recent animal study showed tumor shrinkage when amygdalin was administered at 50 mg/kg—well above safe levels for humans. High doses may cause cyanide toxicity. If used despite warnings, only use food-grade bitter apricot seeds and start with very low amounts. Avoid if pregnant, and avoid co-administration with high-dose vitamin C or probiotics without guidance.

References:

Artemisinin

Simple Summary: Artemisinin stays quiet until it meets iron—then it unleashes ROS that can kill cancer cells by ferroptosis and apoptosis. It also turns down AKT growth signaling and can sensitize tumors to chemo. Human oncology data are still early; artesunate has favorable safety and exposure, especially in short courses or with iron-loading strategies.

Strength of Evidence: ⭐⭐ Preclinical — Strong mechanistic and animal data; early clinical work is emerging but still limited.

Mechanisms: Artemisinin is activated by intracellular iron, triggering cleavage of its endoperoxide bridge and generating reactive oxygen species (ROS). This induces ferroptosis and apoptosis, particularly in cancer cells with high iron uptake. Artemisinin and its derivatives (e.g., artesunate) also inhibit PI3K/AKT/mTOR signaling, can sensitize tumors to chemotherapy, and suppress angiogenesis and metastasis in aggressive cancers.

ROSlab
🔥 Potency7.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.5 / 10
🎯 Translatability6.8 / 10
Endpoint: DCF-DA fluorescence
Anchor: H2O2 100 μM = 10
Iron-activated burst (transferrin/CD71-high cells)
Ferroptosislab
🔥 Potency7.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6.5 / 10
Endpoint: GPX4/lipid-ROS; rescue by ferrostatin-1
Anchor: RSL3 1 μM = 10
Enhanced by iron loading
AKTlab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: p-AKT (S473)
Anchor: LY294002 10 μM = 10
PI3K/AKT/mTOR down-modulation
Apoptosislab
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6.8 / 10
Endpoint: Annexin V / caspase-3/7
Anchor: Staurosporine 1 μM = 10
Mitochondrial pathway after ROS spike

Note: Best as adjunct for iron-rich tumors (e.g., breast, colorectal); artesunate preferred for clinical use due to better PK. Coordinate with iron status; monitor for anemia/ROS-related fatigue.

References:

Ashwagandha

PreclinicalAnti-inflammatoryApoptosis Induction

Simple Summary: Ashwagandha is an adaptogenic herb that may help reduce fatigue from chemotherapy and has shown promise in lab studies for killing cancer cells and curbing inflammation. One study in breast cancer patients found it improved quality of life during treatment.

Strength of Evidence: ⭐⭐ Moderate — Strong preclinical data plus one supportive human trial for symptom management.

Mechanisms: Ashwagandha (Withania somnifera) and its key compound withaferin A induce apoptosis in cancer cells via ROS generation, mitochondrial disruption, and p53 activation. It inhibits NF-κB signaling, reducing inflammation and angiogenesis. Preclinical data show cytotoxicity against breast, lung, colon, and other cancers, with immunomodulatory effects enhancing T-cell activity. In humans, it may alleviate chemotherapy-induced fatigue.

Anti-inflammatorypreclinical
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability7 / 10
Endpoint: NF-κB inhibition / IL-6 ↓
Anchor: Curcumin (clinical) = 10
Withaferin A targets inflammatory pathways in cancer models
Apoptosis Inductionlab
🔥 Potency7.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.5 / 10
🎯 Translatability6.5 / 10
Endpoint: ROS / p53 activation; caspase-3
Anchor: Withaferin A (in vitro) = 10
Selective for cancer cells; mitochondrial pathway

Note: Predominantly preclinical evidence for direct anti-cancer effects; the key human study (PMID 23142798) was an open-label trial in 100 breast cancer patients receiving 2 g of extract three times daily with chemotherapy, showing reduced fatigue and improved quality of life. No large RCTs for anti-tumor efficacy.

References:

Astragaloside IV

PreclinicalApoptosis InductionAnti-metastatic

Simple Summary: Astragaloside IV is a compound from the astragalus plant that triggers cancer cell death and may prevent tumor spread in lab studies. It could make chemotherapy more effective against lung and colon cancers.

Strength of Evidence: ⭐⭐ Moderate — Robust preclinical data; human trials lacking for purified compound.

Mechanisms: Astragaloside IV (AS-IV), a saponin from Astragalus membranaceus, induces apoptosis in cancer cells through mitochondrial disruption, ROS generation, and caspase activation. It inhibits NF-κB signaling to reduce inflammation and angiogenesis, modulates macrophage polarization to curb metastasis, and enhances chemosensitivity by overcoming drug resistance. Preclinical studies demonstrate anti-proliferative effects in lung, colorectal, breast, and other cancers.

Apoptosis Inductionpreclinical
🔥 Potency7.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability6.5 / 10
Endpoint: Caspase-3 / Bcl-2 ↓
Anchor: Withaferin A (in vitro) = 10
Mitochondrial pathway; ROS-dependent
Anti-metastaticlab
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.5 / 10
🎯 Translatability6 / 10
Endpoint: Invasion / migration ↓; MMP-2/9
Anchor: Curcumin (clinical) = 10
Macrophage M2 polarization block

Note: Evidence is predominantly preclinical with in vitro and animal models showing anti-cancer effects. No large-scale human clinical trials for purified AS-IV in cancer; benefits often inferred from astragalus extracts in TCM. Key studies highlight synergy with paclitaxel in breast cancer and cisplatin in lung cancer.

References:

Astragalus Polysaccharide

Early humanTelomereTregsNK Activation

Simple Summary: APS steadies the immune system during treatment—lifting NK function and improving T-cell balance—with human studies showing less fatigue and better tolerance to chemo. Think of it as immune support used alongside standard care; any telomere support is adjunctive, not tumor-killing.

Strength of Evidence: ⭐⭐⭐ Moderate — Human pilot/phase 2 data for fatigue and immune modulation; cancer-control signals mainly from combination formulas; higher-quality trials needed.

Mechanisms: Astragalus polysaccharides (APS, including the injectable PG2) modulate immunity by increasing CD4+ T cells and NK-cell activity, rebalancing Treg/Th17 and Th1/Th2 responses, and helping restore the CD4/CD8 ratio. Via anti-inflammatory signaling (e.g., lower IL-6 and TNF-α), APS has reduced chemotherapy-related fatigue and toxicity in small trials. Astragalus extracts rich in saponins (e.g., astragaloside IV/cycloastragenol) can activate telomerase and support telomere maintenance; this is telomere-related support rather than direct cytotoxicity. Evidence for tumor control is mostly from Astragalus-containing formulas used alongside chemotherapy.

TelomereSupporthuman
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7.5 / 10
Endpoint: hTERT activity / telomere markers
Anchor: Cycloastragenol (TA-65) ref = 10
Adjunctive maintenance; mainly saponin-driven (not APS)
Tregshuman
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity8.0 / 10
🎯 Translatability8 / 10
Endpoint: Treg/Th17; CD4/CD8 balance
Anchor: Metronomic cyclophosphamide (Treg reduction) = 10
Rebalancing rather than depletion
NK Activationhuman
🔥 Potency6.2 / 10
🧪 Confidence4 / 5
🛡 Selectivity8.5 / 10
🎯 Translatability8 / 10
Endpoint: NK cytotoxicity / IFN-γ
Anchor: IL-2 (clinical) = 10
Improved NK function/markers in trials

Note: Human data primarily from PG2 (injectable APS) in phase 2 trials for chemo fatigue (e.g., NCT03441250: 500 mg IV reduced symptoms in solid tumors). Telomere effects from saponin fractions, not APS core; use as adjunct only.

References:

Atorvastatin †Rx

Early humanMevalonateRhoInvasion/MetastasisApoptosis

Simple Summary: A familiar cholesterol drug with anticancer promise: by shutting down the mevalonate pathway, atorvastatin starves tumors of prenylation needed for growth and spread. That can slow migration/metastasis programs and promote apoptosis, and it may enhance chemo or immunotherapy—especially in resistant tumors.

Strength of Evidence: ⭐⭐⭐ Moderate — Human data in ovarian/breast and observational signals across cancers; most robust as an adjunct, with promise in resistant disease.

Mechanisms: Atorvastatin inhibits HMG-CoA reductase, blocking the mevalonate pathway and depleting isoprenoids (FPP/GGPP) required to prenylate small GTPases (Rho/Rac/Ras). Loss of prenylation impairs membrane localization and signaling that drive proliferation, survival, migration, invasion, and therapy resistance. Downstream, this can reduce NF-κB/AKT signaling, lower MMPs/adhesion, curb angiogenesis, and tip cells toward apoptosis. Clinical interest is strongest in drug-resistant disease and as an adjunct to DNA-damaging agents and immunotherapy.

Mevalonatehuman
🔥 Potency8.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability9.5 / 10
Endpoint: HMG-CoA reductase / isoprenoid pool
Anchor: Lovastatin 10 μM = 10
Class effect; high human achievability
Rho (prenylation)human
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability9.5 / 10
Endpoint: Rho/ROCK signaling; migration
Anchor: Y-27632 10 μM (ROCK) = 10
Indirect via geranylgeranyl depletion
Metastasishuman
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability9 / 10
Endpoint: Invasion/adhesion assays; MMPs
Anchor: Marimastat 1 μM = 10
Motility/adhesion suppression
Apoptosislab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability9 / 10
Endpoint: Annexin V / caspase-3/7
Anchor: Staurosporine 1 μM = 10
Context- and lineage-dependent

Note: Lipophilic statin (good tissue penetration, including brain). Signals of synergy have been reported with: Metformin (complementary metabolic/mTOR effects), and Checkpoint inhibitors (possible tumor-microenvironment effects). Potential with Disulfiram (redox/copper toxicity); monitor interactions. All oncology uses require clinician oversight.

References:

Baicalein

PreclinicalNF-κBAutophagyEMTAngiogenesisInflammationMMP

Simple Summary: A multitarget plant flavonoid: baicalein quiets NF-κB/COX-2 inflammatory survival signals, provokes tumor-cell autophagy, and helps block EMT and invasion (MMP-2/9, angiogenesis). Evidence is promising but largely preclinical; achieving active levels usually needs concentrated extracts of baicalein (not just whole-herb).

Strength of Evidence: ⭐⭐ Preliminary — Strong preclinical signals across models; limited human data.

Mechanisms: Baicalein is a flavonoid (5,6,7-trihydroxyflavone) enriched in Scutellaria baicalensis. It suppresses NF-κB and COX-2–linked inflammatory signaling, lowering cytokines (e.g., IL-6, TNF-α) and survival pathways. It can trigger autophagy (↑ Beclin-1/LC3-II; increased flux) and inhibit epithelial–mesenchymal transition (EMT) by rebalancing E-cadherin/vimentin and reducing invasion. Additional actions include downregulation of MMP-2/MMP-9 and anti-angiogenic effects, which together may curb migration/metastasis. Most data are preclinical; concentrated, purified preparations are typically required for pharmacologic effects.

NF-κBlab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: p65 nuclear translocation
Anchor: BAY 11-7082 5 μM = 10
Inflammatory gene suppression
Autophagylab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: LC3-II / Beclin-1; flux
Anchor: Rapamycin 100 nM = 10
Cytostatic/cytotoxic context varies
EMTlab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability6 / 10
Endpoint: Vimentin/E-cadherin; invasion
Anchor: SB431542 10 μM = 10
MMP-2/9, migration ↓

Note: Signals of synergy have been described with: Curcumin (convergent NF-κB/cytokines), EGCG (EMT/CSC programs), and cisplatin (chemosensitization in models).

References:

Baicalin †Botanical

Simple Summary: Baicalin—and its active form, baicalein—turns down survival/growth switches (NF-κB/STAT3 and PI3KmTOR), nudges tumor cells toward apoptosis, and reduces invasiveness by reversing EMT and lowering VEGF/MMPs. It may also reduce PD-L1 in tumor cells and blunt IL-6→STAT3. Orally, parent baicalin is modest; effects likely come from flexible conversion to baicalein and back.

Strength of Evidence: ⭐⭐ Low to Moderate — Robust mechanistic & in vivo signals; limited human oncology outcomes; PK constraints.

Mechanisms: Baicalin (baicalein 7-O-glucuronide) is hydrolyzed by gut β-glucuronidase to baicalein, absorbed, and largely re-glucuronidated with enterohepatic recycling. Across models, baicalin/baicalein suppress NF-κB and JAK/STAT3 signaling, downshift PI3K/AktmTOR growth pathways, and induce mitochondrial apoptosis (Bax↑/Bcl-2↓, caspase-3/PARP cleavage). They curb angiogenesis and invasion via VEGF and MMP-2/9 reduction, reverse EMT (E-cadherin↑; vimentin/Slug↓; TGF-β/Smad3 blockade), and modulate autophagy in a context-dependent fashion. Immune-relevant effects include lowering tumor PD-L1 (e.g., IFN-γ–induced) and dampening IL-6→STAT3 signaling. Oral parent baicalin is low; activity likely reflects dynamic interconversion with baicalein and conjugates.

NF-κBlab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: p65 nuclear translocation
Anchor: BAY 11-7082 5 μM = 10
Inflammation/survival genes ↓
STAT3lab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: p-STAT3 (Y705) / reporter
Anchor: Stattic 5 μM = 10
IL-6 axis attenuation
PI3K/Aktlab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability6 / 10
Endpoint: p-AKT (S473)
Anchor: LY294002 10 μM = 10
mTORlab
🔥 Potency5.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability6 / 10
Endpoint: p-mTOR / S6K
Anchor: Rapamycin 100 nM = 10
Apoptosislab
🔥 Potency5.6 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: Caspase-3/7 / Annexin V
Anchor: Staurosporine 1 μM = 10
EMTlab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability6 / 10
Endpoint: E-cadherin↑ / Vimentin↓
Anchor: SB431542 10 μM = 10
VEGFlab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: VEGF / tube formation
Anchor: Sunitinib 50–100 nM = 10
MMP-2/9lab
🔥 Potency5.7 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: Zymography / invasion
Anchor: Marimastat 1 μM = 10
PD-L1lab
🔥 Potency4.8 / 10
🧪 Confidence2 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: PD-L1 protein/mRNA
Anchor: Ruxolitinib 1 μM (IFN-γ PD-L1 block) = 10
Autophagylab
🔥 Potency4.8 / 10
🧪 Confidence2 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability6 / 10
Endpoint: LC3-II / flux
Anchor: Rapamycin 100 nM = 10
Context-dependent

Note: Best fit in protocols aiming to dampen NF-κB/STAT3 and PI3KmTOR, reduce EMT/angiogenesis, and potentially aid radio/chemo-sensitization. Human oncology outcome data are limited; consider bioavailability and potential UGT/CYP modulation.

References:

Beetroot Powder

PreclinicalNO DonorPerfusionOxidative Stress Modulation

Simple Summary: By raising nitric oxide, beetroot can widen blood vessels and improve tissue oxygenation—potentially helping radiation and some drugs in hypoxic tumors. Its pigments (betalains) add antioxidant/anti-inflammatory support. Consider it as supportive care; anticancer effects are mostly preclinical.

Strength of Evidence: ⭐⭐ Preliminary — Human physiology data for perfusion; anticancer signals mainly preclinical.

Mechanisms: Beetroot is rich in dietary nitrates that convert to nitric oxide (NO), a vasodilator that can increase tissue blood flow and oxygenation. Improved oxygen delivery may enhance sensitivity to radiation and some chemotherapies in hypoxic tumors. Beetroot’s betalain pigments have strong antioxidant and anti-inflammatory activity, reducing lipid peroxidation and related signaling. NO donors also modulate tumor biology, including immune responses, in preclinical models.

NO Donorhuman
🔥 Potency5.8 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability9 / 10
Endpoint: Plasma NOx / cGMP / BP
Anchor: Sodium nitroprusside = 10
Physiologic NO rise (dietary route)
Perfusionhuman
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability9 / 10
Endpoint: Tissue pO₂/NIRS/BF
Anchor: Pharmacologic vasodilator = 10
Greatest in hypoxia/exercise
Oxidative StressModulationlab
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability8 / 10
Endpoint: Lipid peroxidation / antioxidant markers
Anchor: N-acetylcysteine = 10
Antioxidant bias—time away from ROS-based hits

Note: Possible synergies: radiation (better oxygenation → radiosensitivity), HBOT/high-O₂ strategies, and exercise with dietary nitrate to boost delivery/utilization.

References:

Berberine

Early humanAMPKMetabolismMetastasis

Simple Summary: Berberine flips on AMPK, throttling tumor sugar/fat metabolism and dampening invasion programs. It can raise oxidative stress, impair DNA repair, and re-sensitize resistant ovarian cancer cells to chemo. Human data are strongest for metabolic effects; oncology signals are accumulating.

Strength of Evidence: ⭐⭐⭐ Moderate — Strong preclinical synergies; some human data in metabolic cancers, with ongoing trials for ovarian resistance.

Mechanisms: Berberine, an isoquinoline alkaloid, activates AMPK, suppressing glycolysis and oxidative phosphorylation (OXPHOS), disrupting the Warburg effect. It inhibits lipid metabolism, reducing metastasis via MMP16 downregulation, and induces oxidative DNA damage while impairing homologous recombination repair. In ovarian cancer, it modulates autophagy through the LINC01123/p65/MAPK10 axis and sensitizes cells to chemotherapy by overcoming resistance mechanisms.

AMPKhuman
🔥 Potency7.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability8.5 / 10
Endpoint: p-AMPK (Thr172), ACC phosphorylation
Anchor: AICAR/Metformin (clinical) = 10
Downstream mTOR/glycolysis suppression
Metabolism↓ (glycolysis/OXPHOS)human
🔥 Potency6.8 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability8.5 / 10
Endpoint: ECAR/OCR, lactate output, lipid synthesis
Anchor: 2-DG 5 mM (glycolysis block) = 10
Warburg effect dampened; lipogenesis↓
Metastasislab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: MMP-16, migration/invasion assays
Anchor: Marimastat 1 μM = 10
Adhesion/matrix programs reduced

Note: Potential partners: Curcumin (AMPK↑/apoptosis↑), Resveratrol (mTOR↓; anti-metastatic), Quercetin (DNA damage/autophagy), PARP inhibitors (HR-deficient tumors), Cisplatin (resistance reversal), Alpelisib (PI3K). Watch for drug interactions (P-gp/CYP3A4) and GI intolerance at higher doses.

References:

Betulinic Acid

PreclinicalMitoROSAngiogenesis

Simple Summary: A triterpenoid from birch bark, betulinic acid collapses tumor mitochondria, spikes ROS, and reduces new blood-vessel growth. Preclinical work—especially in ovarian cancer—shows apoptosis and G2/M arrest, with hints of immune reprogramming (M2→M1). Delivery tech (nanoformulations) is being explored to improve exposure.

Strength of Evidence: ⭐⭐ Low to Moderate — Strong preclinical data in ovarian and other cancers; nano-formulations in early trials, but limited human studies.

Mechanisms: Betulinic acid induces mitochondrial apoptosis through a ROS burst, cytochrome c release, and caspase activation. It inhibits angiogenesis by downregulating VEGF and suppresses topoisomerase I/II, leading to DNA damage. In ovarian cancer models, it promotes apoptosis and cell cycle arrest at G2/M phase by upregulating metallothionein 1G. It also reprograms tumor-associated macrophages from M2 (pro-tumor) to M1 (anti-tumor) phenotype, reducing immunosuppression in the tumor microenvironment.

Mitochondria↑ (apoptosis)lab
🔥 Potency7.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5.5 / 10
Endpoint: Δψm loss (JC-1/TMRM), cytochrome c release
Anchor: CCCP 10 μM (Δψm collapse) = 10
Bax↑/Bcl-2↓ → caspase-9/3 activation
ROSlab
🔥 Potency6.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5 / 10
Endpoint: DCF-DA fluorescence; 8-oxo-dG
Anchor: H2O2 100 μM = 10
ROS burst precedes mitochondrial apoptosis
Angiogenesislab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5 / 10
Endpoint: VEGF/VEGFR2; endothelial tube formation
Anchor: Sunitinib 50–100 nM = 10
Anti-VEGF signaling; invasion markers↓

Note: Promising synergies: Curcumin (NF-κB↓; apoptosis↑), Resveratrol (ROS-mediated apoptosis; anti-angiogenic), Quercetin (DNA damage/cell-cycle arrest), PARP inhibitors (HR-deficient tumors), Cisplatin (overcome resistance), Everolimus (mTOR targeting).

References:

BioBran (MGN-3 / RBAC)

Early humanNKTh1 CytokinesDCQoL

Simple Summary: Primarily an immune adjuvant: BioBran boosts NK function, dendritic cells, and Th1 cytokines. Small trials point to better quality of life and, in one liver-cancer RCT, lower recurrence and improved survival when added to standard therapy.

Strength of Evidence: ⭐⭐⭐ Moderate — Small RCTs/pilots show immune modulation and QoL benefits; one randomized liver-cancer trial suggests reduced recurrence and better survival with adjunct MGN-3. Larger confirmatory trials are needed.

Mechanisms: BioBran (modified rice bran arabinoxylan) acts as a biological response modulator. In human studies it increases natural killer (NK) cell cytotoxicity, raises myeloid dendritic cells, and shifts cytokines toward a Th1 profile (↑IFN-γ, ↑IL-12, ↑TNF-α), enhancing antitumor immune surveillance. In a randomized clinical trial in hepatocellular carcinoma, adding MGN-3 to interventional therapy reduced recurrence, lowered AFP, and was associated with better 2-year survival; pilot trials suggest improved quality of life during chemotherapy. Preclinical work shows pro-apoptotic signaling and synergy with chemotherapy, but BioBran is primarily immunomodulatory rather than directly cytotoxic.

NKhuman
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity8.5 / 10
🎯 Translatability8.5 / 10
Endpoint: NK cytotoxicity (% kill), IFN-γ
Anchor: IL-2 (clinical) = 10
Multiple trials show NK function↑
Th1 Cytokineshuman
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.5 / 10
🎯 Translatability8 / 10
Endpoint: IL-12/IFN-γ/TNF-α levels
Anchor: IL-12 therapy = 10
Th1 bias with safety
Dendritic Cellshuman
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity7.5 / 10
🎯 Translatability8 / 10
Endpoint: mDC counts; HLA-DR/CD86
Anchor: GM-CSF (clinical DC induction) = 10
Improved antigen presentation markers
Quality of Lifehuman
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8 / 10
Endpoint: FACT-G / EORTC QLQ-C30
Anchor: FACT-G +10 points = 10
Pilot benefits during chemotherapy

Note: Generally well-tolerated; best as an adjunct. Use caution with immunosuppression or transplants.

References:

Black Seed Oil / Thymoquinone

PreclinicalPI3KROSApoptosis

Simple Summary: From black cumin seed, thymoquinone turns down PI3K/AKT survival signaling, raises ROS in tumor cells, and triggers apoptosis. It shows synergy with standard drugs (e.g., cisplatin, doxorubicin) in preclinical ovarian models.

Strength of Evidence: ⭐⭐ Low to Moderate — Robust preclinical data, including ovarian synergies; limited human trials, but promising for adjunctive use.

Mechanisms: Thymoquinone (TQ), the active compound in black seed oil, inhibits PI3K/Akt and STAT3 pathways, blocking cancer cell survival and proliferation. It enhances TRAIL-mediated apoptosis, increases ROS selectively in cancer cells, and induces cell cycle arrest. In ovarian cancer, TQ promotes apoptosis via p53-dependent mechanisms, reduces NF-κB activity, and sensitizes cells to chemotherapy by targeting resistance pathways.

PI3K/AKTlab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: p-AKT (S473) / downstream
Anchor: LY294002 10 μM = 10
STAT3 and NF-κB also ↓ in models
ROSlab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability6 / 10
Endpoint: DCF-DA; mitochondrial ROS
Anchor: H2O2 100 μM = 10
Pro-oxidant bias in cancer cells
Apoptosislab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability6 / 10
Endpoint: Caspase-3/7; Annexin V
Anchor: Staurosporine 1 μM = 10
TRAIL sensitization; p53-dependent signals

Note: Acts antioxidant in normal cells yet pro-oxidant in tumors. Synergies reported with curcumin, berberine, resveratrol, cisplatin, doxorubicin; potential immune-modulating benefits are under study.

References:

Black Walnut / Juglone

PreclinicalROSEMTMMP

Simple Summary: Juglone overwhelms tumor cells with ROS, poisons DNA copying/repair, and downshifts invasion programs (EMT/MMPs). It’s potent in dishes and animals, but human trials are lacking and liver toxicity is a concern—so it’s exploratory, not clinic-ready.

Strength of Evidence: ⭐⭐ Low — Robust preclinical data across multiple cancers; no human trials, with concerns about hepatotoxicity limiting clinical use.

Mechanisms: Juglone, a naphthoquinone from black walnut (Juglans nigra), generates reactive oxygen species (ROS) via redox cycling, inducing oxidative stress and apoptosis in cancer cells. It inhibits topoisomerase-II, disrupting DNA replication and repair, and targets Pin1, a prolyl isomerase, to block cancer cell proliferation and metastasis. It also modulates p53 and STAT3 pathways, enhancing cell cycle arrest and reducing tumor invasion.

ROSlab
🔥 Potency7.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability4.5 / 10
Endpoint: DCF-DA / MitoSOX
Anchor: H2O2 100 μM = 10
Redox cycling quinone; toxicity limits dosing window
EMTlab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4.5 / 10
Endpoint: E-cadherin↑ / Vimentin↓ / Slug↓
Anchor: SB431542 10 μM = 10
Via Pin1/STAT3/NF-κB modulation
MMP (2/9)lab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4.5 / 10
Endpoint: Gelatin zymography / invasion
Anchor: Marimastat 1 μM = 10
Anti-invasion; model-dependent

Note: Potential hepatotoxicity—medical supervision required. Studied in breast, lung, liver, pancreatic cancers and leukemia. Reported synergies: curcumin (NF-κB/ROS), berberine (ROS/STAT3), resveratrol (DNA damage), doxorubicin, cisplatin, etoposide.

References:

Boswellia / AKBA

Early human5-LOXNF-κBApoptosis

Simple Summary: AKBA turns down leukotriene/NF-κB inflammation, switches on apoptosis, and lowers invasion/angiogenesis genes. Human signals include less brain-tumor edema and reduced tumor proliferation in a breast ‘window’ trial; direct survival benefits remain unproven.

Strength of Evidence: ⭐⭐ Low to Moderate — Strong mechanistic and in vivo data; early human signals (breast window trial, brain-tumor edema) but limited cancer-outcome trials.

Mechanisms: Acetyl-11-keto-β-boswellic acid (AKBA) directly inhibits 5-lipoxygenase, lowering leukotrienes and inflammation; suppresses NF-κB–regulated survival, proliferation, and angiogenic genes; inhibits STAT3 signaling; and induces caspase-dependent apoptosis. In vivo, AKBA reduces tumor growth and metastasis in a pancreatic cancer model with downregulation of VEGF, COX-2, MMP-9, and CXCR4. AKBA can reach the brain in animals, boswellic acids show activity against glioblastoma cells, and Boswellia extracts have reduced cerebral edema in brain-tumor patients and decreased proliferation in a breast cancer window trial.

5-LOXhuman
🔥 Potency7.8 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability8 / 10
Endpoint: Leukotrienes (LTB4) / 5-HETE
Anchor: Zileuton clinical effect = 10
Direct enzymatic inhibition by AKBA
NF-κBlab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: p65 nuclear translocation / reporters
Anchor: BAY 11-7082 5 μM = 10
Survival/angiogenic genes down
Apoptosislab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: Caspase-3/7; Annexin V
Anchor: Staurosporine 1 μM = 10
Context-dependent across lineages

Note: Use cautiously with anticoagulants/antiplatelets. Most oncology efficacy data are preclinical; clinical signals are supportive (edema/proliferation markers).

References:

Brazil Nuts / Selenium

ClinicalGPxTrxRApoptosis

Simple Summary: Brazil nuts raise selenium and boost antioxidant enzymes (GPx, TrxR) in humans; selenium metabolites can trigger tumor-cell apoptosis in models. Benefits are dose- and baseline-dependent, and prevention trials are mixed. Use within a safe window to avoid selenosis.

Strength of Evidence: ⭐⭐⭐⭐ Strong — Human RCTs show Brazil nuts elevate Se and GPx; oncology prevention/therapy data are mixed and status-dependent.

Mechanisms: Selenium, abundant in Brazil nuts, upregulates selenoproteins like glutathione peroxidase (GPx) and thioredoxin reductase (TrxR), reducing oxidative stress and neutralizing free radicals. Methylselenol, a selenium metabolite, triggers p53-mediated apoptosis and inhibits tumor progression. Selenium status also shapes immune function, enhancing NK- and T-cell activity in supplementation studies. Selenium is central to the GPX4 pathway that suppresses ferroptosis; selenium deprivation can induce ferroptosis, a mechanism relevant to tumor progression and metastasis biology.

GPxhuman
🔥 Potency8.2 / 10
🧪 Confidence5 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability9.5 / 10
Endpoint: GPx activity (erythrocyte/plasma)
Anchor: Selenomethionine 200 μg/day = 10
Robust rise with 1–2 Brazil nuts in low-Se populations
TrxRhuman
🔥 Potency6.2 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability9 / 10
Endpoint: Thioredoxin reductase activity
Anchor: Selenite 200 μg/day = 10
Selenoprotein upregulation; tissue-specific variability
Apoptosislab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability6 / 10
Endpoint: Caspase-3/7; p53 activation
Anchor: Staurosporine 1 μM = 10
Driven by methylselenol/selenite in models; dose window narrow in humans

Note: Narrow therapeutic window (~55–400 μg/day total intake). Excess → selenosis (hair/nails, GI), and some trials signal diabetes risk. Mixed prevention data (e.g., SELECT null; NPH trial skin cancer signal). Synergies under study include combos with targeted therapies (e.g., axitinib in RCC).

References:

CBD (Cannabidiol)

PreclinicalApoptosisNF-κBAngio

Simple Summary: Non-intoxicating CBD can push cancer cells toward apoptosis, dial down NF-κB inflammation, and reduce new blood-vessel growth in lab models. Early human oncology data are limited; watch for drug interactions (strong CYP2C19/3A4 inhibition) and liver-enzyme elevations at higher doses.

Strength of Evidence: ⭐⭐ Preclinical — Strong cell/animal data; limited human trials but promising for adjunctive therapy.

Mechanisms: CBD activates TRPV1 and PPAR-γ receptors, reducing tumor cell proliferation and inducing apoptosis via caspase activation. It downregulates Id-1, a metastasis-promoting gene, and inhibits angiogenesis by suppressing VEGF and MMPs. CBD also reduces NF-κB signaling, decreasing inflammation-driven tumor growth, and modulates CB1/CB2 receptors to disrupt cancer cell migration. Emerging evidence suggests CBD enhances chemotherapy and radiation therapy efficacy by promoting autophagic cell death and overcoming resistance.

Apoptosislab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability6 / 10
Endpoint: Caspase-3/7; Annexin V
Anchor: Staurosporine 1 μM = 10
TRPV1/PPAR-γ mediated; Id-1 ↓ contributes
NF-κBlab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: p65 nuclear translocation / reporter
Anchor: BAY 11-7082 5 μM = 10
Inflammation/survival genes ↓
Angiogenesislab
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5.5 / 10
Endpoint: VEGF/MMPs; endothelial tube formation
Anchor: Sunitinib 50–100 nM = 10
Id-1 ↓ correlates with metastasis traits

Note: CYP interactions: may raise levels of chemo, TKIs, azoles, warfarin, antiepileptics—coordinate with oncology/pharmacy. Common AEs: somnolence, diarrhea, appetite change, transaminitis (dose-related). Signals of synergy have been reported with: Chemotherapy (e.g., TMZ, cisplatin - enhanced apoptosis and resistance reversal), Radiation (potentiates effects), Curcumin (complementary anti-inflammatory/apoptosis). All oncology uses require clinician oversight.

References:

Chlorella Powder

Early humanDetoxNKApoptosis

Simple Summary: Chlorella helps block/clear some food-borne carcinogens, boosts NK-cell activity in human trials, and can trigger tumor-cell apoptosis in lab work. It’s best viewed as preventive/supportive—pairing with therapy and diet cleanup—rather than a stand-alone anticancer treatment.

Strength of Evidence: ⭐⭐⭐ Moderate — Human trials show immune/antioxidant effects and reduced carcinogen exposure; anti-tumor actions mostly preclinical with limited patient data.

Mechanisms: Chlorella’s chlorophyllin binds dietary carcinogens (e.g., aflatoxin, heterocyclic amines), reducing their bioavailability. Supplementation can increase NK cell activity and Th1 cytokines (IFN-γ, IL-12) in humans, upregulate antioxidant enzymes (SOD, catalase), and in cancer cell models trigger apoptosis via p53/Bax/caspase-3 with decreased Bcl-2.

Detox (carcinogen binding)human
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8.5 / 10
Endpoint: Aflatoxin–DNA adducts; urinary HCA markers
Anchor: Chlorophyllin clinical effect = 10
Diet-dependent; works best when exposure present
NKhuman
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity8.0 / 10
🎯 Translatability8.5 / 10
Endpoint: NK cytotoxicity; IFN-γ/IL-12
Anchor: IL-2 (clinical) = 10
Th1 bias seen in RCTs
Apoptosislab
🔥 Potency4.5 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5 / 10
Endpoint: Caspase-3; Bax/Bcl-2; TUNEL
Anchor: Staurosporine 1 μM = 10
Primarily in vitro

Note: Quality varies—choose tested, low-contaminant lots. Vitamin K content may antagonize warfarin. Possible GI upset; rare algae/iodine sensitivities. Immunostimulatory—use caution in autoimmunity or post-transplant. Signals of synergy have been reported with: Spirulina (immune/detox co-boost), Curcumin (anti-inflammatory/apoptosis), Chemotherapy (possible adjunct QoL). All oncology uses require clinician oversight.

References:

CoQ10 (Ubiquinol)

Early humanMito SupportCardio-ProtectApoptosis

Simple Summary: CoQ10 supports mitochondrial energy and helps protect the heart during cardiotoxic therapies; small trials and meta-analyses show benefit signals. In lab models it can push cancer cells toward apoptosis and tone down ERK/AKT/VEGF, but clinical antitumor data are early.

Strength of Evidence: ⭐⭐⭐ Moderate — Human studies support cardioprotection/biomarker changes; antitumor effects mainly preclinical.

Mechanisms: CoQ10, an electron carrier in the mitochondrial electron transport chain (ETC), enhances ATP production and reduces oxidative stress via antioxidant properties. It mitigates anthracycline- and HER2-therapy–related cardiotoxicity by stabilizing mitochondrial membranes. CoQ10 also induces apoptosis in cancer cells and downregulates pro-growth signaling (e.g., ERK/Akt), while reducing angiogenic factors (e.g., VEGF) and inflammatory signaling.

Mito Supporthuman
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability9.5 / 10
Endpoint: ATP/ETC function; plasma/RBC CoQ10 levels
Anchor: High-bioavailability ubiquinol PK = 10
Supports ETC; antioxidant membrane stabilization
Cardio-Protecthuman
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity8.0 / 10
🎯 Translatability9 / 10
Endpoint: LVEF preservation; troponin/BNP
Anchor: Dexrazoxane (clinical) = 10
Signals with anthracyclines/HER2 agents
Apoptosislab
🔥 Potency4.8 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: Caspase-3/7; ERK/AKT ↓; VEGF ↓
Anchor: Staurosporine 1 μM = 10
Context-dependent, mostly preclinical

Note: Generally well-tolerated. May blunt warfarin effect; separate from some TKIs if advised. Forms: ubiquinol (reduced) often has better bioavailability than ubiquinone at equal mg. Signals of synergy have been reported with: Anthracyclines (cardioprotection), Metformin (mito/metabolic co-support), Curcumin (antioxidant/apoptosis). All oncology uses require clinician oversight.

References:

Curcumin / Theracurmin

ClinicalNF-κBSTAT3ApoptosisAngio ↓

Simple Summary: Well-studied turmeric extract that shuts down NF-κB/STAT3 inflammation, nudges tumor cells into apoptosis, and reduces VEGF/MMPs. Bioavailability is the bottleneck—formulations like Theracurmin improve exposure. Human trials show signals across several cancers and for chemo-sensitization.

Strength of Evidence: ⭐⭐⭐⭐ Strong — Supported by multiple RCTs and meta-analyses, especially for colorectal and pancreatic cancers.

Mechanisms: Curcumin, a turmeric-derived polyphenol, inhibits NF-κB and STAT3 pathways, reducing inflammation-driven tumor growth and metastasis. It downregulates anti-apoptotic proteins (Bcl-2, Bcl-xL), upregulates pro-apoptotic Bax, and activates caspases, inducing apoptosis. Curcumin resensitizes tumor cells to platinum-based chemotherapy by inhibiting ABC transporters and DNA repair pathways. It also suppresses VEGF, MMP-2, and MMP-9, inhibiting angiogenesis, and modulates PI3K/Akt/mTOR to limit cell proliferation.

NF-κBhuman
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability8 / 10
Endpoint: p65 nuclear translocation / NF-κB reporter
Anchor: BAY 11-7082 5 μM = 10
CRP/IL-6 often ↓ in trials
STAT3lab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: p-STAT3 (Y705); target genes
Anchor: Stattic 5 μM = 10
Apoptosishuman
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability7.5 / 10
Endpoint: Caspase-3/7; Bax↑/Bcl-2↓; TUNEL
Anchor: Staurosporine 1 μM = 10
Tissue biomarker shifts in window studies
Angiogenesislab
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: VEGF/MMP-2/9; tube formation
Anchor: Sunitinib 50–100 nM = 10

Note: Potential CYP/P-gp interactions; separate from some TKIs/chemo per pharmacist. Watch for GI upset and rare LFT elevations. Pairing with piperine increases absorption but also raises interaction risk. Signals of synergy have been reported with: Piperine (bioavailability boost), Resveratrol (NF-κB/STAT3 co-inhibition), Cisplatin (resistance reversal). All oncology uses require clinician oversight.

References:

Dandelion Root

PreclinicalMito CollapseAutophagyPI3KAngio

Simple Summary: In lab models, DRE collapses tumor mitochondrial potential, triggers autophagy-linked death, and tones down PI3K/VEGF signaling. Human oncology data are limited; treat as an adjunct under supervision.

Strength of Evidence: ⭐⭐ Preclinical — Strong in vitro and animal data; human trials are limited but approved, with recruitment challenges.

Mechanisms: Dandelion root extract (DRE) induces mitochondrial depolarization, disrupting membrane potential and releasing cytochrome c, leading to caspase-mediated apoptosis. It upregulates Beclin-1 and LC3-II to promote autophagic cell death. DRE inhibits PI3K/Akt signaling, reducing proliferation, and downregulates VEGF to suppress angiogenesis. Preclinical studies show selective toxicity to cancer cells, sparing healthy cells.

Mitochondrial ΔψmCollapselab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5.5 / 10
Endpoint: Δψm loss (JC-1/TMRM); cytochrome c release
Anchor: CCCP 10 μM = 10
Autophagylab
🔥 Potency5.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5.5 / 10
Endpoint: LC3-II; Beclin-1; flux assays
Anchor: Rapamycin 100 nM = 10
PI3K/AKTlab
🔥 Potency5.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability5 / 10
Endpoint: p-AKT (S473); downstream
Anchor: LY294002 10 μM = 10
Angiogenesislab
🔥 Potency4.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.5 / 10
🎯 Translatability4.5 / 10
Endpoint: VEGF; endothelial tube formation
Anchor: Sunitinib 50–100 nM = 10

Note: Possible ragweed-family cross-reactivity; mild diuresis; GI upset. Standardize extracts if possible; avoid if on potent diuretics without guidance. Signals of synergy have been reported with: Curcumin (autophagy/apoptosis co-boost), Berberine (PI3K/Akt inhibition), Chemotherapy (potential sensitization). All oncology uses require clinician oversight.

References:

DCA (Dichloroacetate Sodium)

ClinicalPDKMitoApoptosisHIF-1α

Simple Summary: By blocking PDK, DCA forces tumors to burn fuel in mitochondria again—raising ROS and tipping cells into apoptosis. Human pilot data exist; it can help sensitize to chemo/radiation. Requires clinician oversight due to neuropathy risk.

Strength of Evidence: ⭐⭐⭐ Moderate — Supported by human pilot studies and strong mechanistic data; RCTs are limited.

Mechanisms: DCA inhibits pyruvate dehydrogenase kinase (PDK), reactivating pyruvate dehydrogenase to shift cancer cell metabolism from glycolysis to oxidative phosphorylation, countering the Warburg effect. This elevates mitochondrial ROS, triggering caspase-3-mediated apoptosis. DCA downregulates Bcl-2, enhances p53 activity, and inhibits HIF-1α, reducing angiogenesis. It sensitizes tumors to chemotherapy and radiation.

PDKhuman
🔥 Potency8.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability8.5 / 10
Endpoint: PDH E1α de-phosphorylation (Ser293); PDH activity
Anchor: AZD7545 10 μM = 10
Mitochondrial Respirationhuman
🔥 Potency7.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability8 / 10
Endpoint: OCR (Seahorse), lactate↓/pyruvate↑
Anchor: Maximal OCR restoration = 10
Apoptosislab
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: Caspase-3/7; Bcl-2↓; p53↑
Anchor: Staurosporine 1 μM = 10
HIF-1αlab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: HIF-1α protein/mRNA; VEGF
Anchor: Acriflavine 5 μM = 10

Note: †Use with medical supervision. Common dose-limiter: peripheral neuropathy; monitor LFTs and B12/thiamine status. Consider pulsed schedules and co-factors (e.g., ALA) when appropriate. Signals of synergy have been reported with: Platinum chemo (resistance reversal), Radiation (ROS boost), Metformin (metabolic co-targeting). All oncology uses require clinician oversight.

References:

Disulfiram + Cu †Rx

Early humanALDHProteasomeCSC

Simple Summary: Repurposed alcoholism drug + copper that blocks protein disposal and hits ALDH-high cancer stem cells. It can heighten chemo/radiation effects. Human signals exist (mixed across cancers), so use is investigational and requires close MD oversight.

Strength of Evidence: ⭐⭐⭐ Moderate — Early-phase human trials and strong preclinical data; mixed outcomes in larger studies (e.g., GBM).

Mechanisms: Disulfiram (DSF), when combined with copper (Cu), forms the Cu-DDC (copper-diethyldithiocarbamate) complex, which potently inhibits proteasome activity, disrupting protein degradation and inducing endoplasmic reticulum stress in cancer cells. It suppresses NF-κB signaling, reducing inflammation-driven tumor growth and metastasis. DSF/Cu targets cancer stem cells (CSCs) by inhibiting aldehyde dehydrogenase (ALDH), increasing reactive oxygen species (ROS), and activating caspase-mediated apoptosis. It also enhances chemotherapy and radiation sensitivity by impairing DNA repair pathways.

ALDHhuman
🔥 Potency7.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: ALDEFLUOR+ fraction; ALDH activity
Anchor: DEAB 50 μM = 10
CSC marker suppression aligns with tumorsphere↓
Proteasomelab
🔥 Potency7.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6.5 / 10
Endpoint: Chymotrypsin-like activity; ubiquitinylated protein↑
Anchor: Bortezomib 10 nM = 10
Cu-DDC drives ER-stress/apoptosis
CSClab
🔥 Potency6.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.5 / 10
🎯 Translatability6.5 / 10
Endpoint: Tumorsphere formation; CD44+/ALDHhigh frequency
Anchor: Salinomycin 1 μM = 10
Stem-like subpopulations reduced

Note: †Use with medical supervision. Strict alcohol avoidance (disulfiram–ethanol reaction). Monitor LFTs and neuropathy. Copper co-administration typically low-dose; avoid copper overload. Drug–drug interactions via CYP metabolism are possible. Signals of synergy have been reported with: Platinum chemo (resistance reversal), Radiation (ROS boost), Curcumin (ROS/apoptosis boost), Resveratrol (NF-κB inhibition), EGCG (CSC targeting), Quercetin (proteasome inhibition). All oncology uses require clinician oversight.

References:

DMF (Dimethyl Fumarate) †Rx

PreclinicalNRF2ROSNF-κB

Simple Summary: MS drug that turns on the NRF2 defense program while paradoxically pushing over-stressed cancer cells into ROS-driven death. Strong lab support; clinical cancer trials are not yet established.

Strength of Evidence: ⭐⭐ Preclinical — Robust mechanistic/animal data; no human oncology trials yet.

Mechanisms: DMF alkylates Keap1 to activate NRF2, inducing antioxidant/phase-II enzymes (HO-1, NQO1). Tumor cells, already redox-stressed, can tip into lethal ROS with DMF. DMF also inhibits NF-κB and downshifts PI3K/AKT, promoting apoptosis in multiple preclinical models.

NRF2lab
🔥 Potency8.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: ARE-luciferase; HO-1/NQO1 induction
Anchor: CDDO-Me 30 nM = 10
Keap1 cysteine succination
ROS↑ (in tumors)lab
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.5 / 10
🎯 Translatability6 / 10
Endpoint: DCF-DA; mitochondrial ROS
Anchor: H2O2 100 μM = 10
Context-dependent; normal cells buffered by NRF2
NF-κBlab
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: p65 nuclear translocation; reporter
Anchor: BAY 11-7082 5 μM = 10
Inflammatory/proliferative genes ↓

Note: †Use with medical supervision. On-label monitoring applies off-label: lymphopenia (check CBC), LFTs, flushing/GI effects. Consider infection risk with low lymphocytes. Coordinate timing with other redox-active agents. DMF is FDA-approved for multiple sclerosis but requires medical supervision for off-label cancer use due to risks of lymphopenia, gastrointestinal issues, and flushing. Studied in: glioblastoma, breast, and colorectal cancers. Synergies include: Curcumin (Natural): Enhances ROS-induced apoptosis in glioblastoma. Radiotherapy: Increases tumor cell sensitivity in breast cancer. Temozolomide (Rx): Synergizes to enhance apoptosis in glioblastoma. Resveratrol (Natural): Boosts NRF2 activation and ROS stress in colorectal cancer. Green Tea Extract (EGCG) (Natural): Enhances NF-κB inhibition and apoptosis in breast cancer. Quercetin (Natural): Synergizes to increase ROS and inhibit PI3K/Akt in glioblastoma.

References:

Doxycycline †Rx

Early humanMMPCSCMito

Simple Summary: An antibiotic that targets tumor mitochondria (especially in stem-like cells) and blocks MMPs that tumors use to invade. Human pilot data exist; larger trials are needed.

Strength of Evidence: ⭐⭐⭐ Moderate — Early human data plus strong mechanistic support; confirmatory RCTs pending.

Mechanisms: Doxycycline inhibits mitochondrial ribosomes, reducing OXPHOS and preferentially impairing cancer stem cells. It also inhibits MMP-2/-9 to reduce invasion/metastasis, can lower drug efflux (e.g., P-gp), and triggers ROS-linked apoptosis.

MMP-2/9human
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.5 / 10
🎯 Translatability7.5 / 10
Endpoint: Gelatin zymography; invasion assays
Anchor: Marimastat 1 μM = 10
Anti-invasion/metastasis
CSClab
🔥 Potency6.2 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6.5 / 10
Endpoint: Tumorspheres; ALDHhigh/CD44+ fractions
Anchor: Salinomycin 1 μM = 10
Mito-dependency of CSCs exploited
Mitochondrial Translation/OXPHOSlab
🔥 Potency6.8 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: mt-protein synthesis; OCR (Seahorse)
Anchor: Tigecycline 5 μM = 10
Leads to ROS↑ and apoptosis

Note: †Use with medical supervision. Photosensitivity, GI upset, esophagitis risk; avoid with retinoids; separate from polyvalent cations. Long courses raise antimicrobial-resistance concerns—use only with oncology guidance. Requires medical supervision due to risks of photosensitivity, gastrointestinal upset, and antibiotic resistance. Studied in: breast, pancreatic, lung, and colorectal cancers. Synergies include: Metformin (Rx): Enhances CSC targeting in breast cancer. Chemotherapy (e.g., Paclitaxel): Increases tumor sensitivity in lung cancer. Berberine (Natural): Synergizes to inhibit MMPs in colorectal cancer. Resveratrol (Natural): Boosts CSC targeting and apoptosis in breast cancer. Green Tea Extract (EGCG) (Natural): Enhances ROS and chemotherapy sensitivity in lung cancer. Quercetin (Natural): Synergizes to inhibit MMPs and induce apoptosis in pancreatic cancer.

References:

EGCG (Epigallocatechin Gallate)

ClinicalPI3KDNMTMMPVEGF

Key Takeaway: Green tea extract that may turn tumor-suppressing genes back on and block blood supply to tumors, while slowing cancer growth and triggering cell death, with evidence from human trials.

Strength of Evidence: ⭐⭐⭐⭐ Moderate to Strong — Strong preclinical/mechanistic data, Moderate clinical data. Supported by multiple RCTs and meta-analyses, especially for colorectal and lung cancers.

Mechanisms: EGCG, the primary catechin in green tea, inhibits PI3K/Akt/mTOR signaling, reducing cell proliferation and survival. It acts as a DNMT inhibitor, demethylating tumor suppressor genes (e.g., p16, MGMT) to restore their expression. EGCG suppresses MMP-2 and MMP-9, limiting tumor invasion and metastasis, and downregulates VEGF to inhibit angiogenesis. It also induces apoptosis by upregulating Bax, activating caspases, and modulating NF-κB pathways. Clinical studies show enhanced chemotherapy efficacy.

PI3K/Akt/mTORHuman biomarkers + RCT adjunct; strong preclinical
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: p-Akt (Ser473) reduction (% vs control) @4–24h in tumor cells at ≤20 µM
Anchor: 10 = LY294002 10 µM or MK-2206 1 µM → ≥90% p-Akt reduction in same assay
Effective concentrations in vitro often ≥5–20 µM; human Cmax with standard extracts is low. Use translatability to reflect PK limits; nano/optimized EGCG may improve.
DNMT (epigenetic)In vitro demethylation; limited clinical epigenetic readouts
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: Promoter methylation change of p16/MGMT (Δ% methylation) and re-expression after ≤20–50 µM EGCG
Anchor: 10 = Decitabine (or 5-aza-dC) 1 µM → robust demethylation/re-expression in same model
EGCG demethylation is weaker vs nucleoside DNMT inhibitors and often needs higher, less clinically achievable doses.
MMP-2/9 (invasion)Human surrogate endpoints + preclinical
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: MMP-2/9 activity by gelatin zymography (% reduction) and invasion assays at ≤10–20 µM
Anchor: 10 = Marimastat 1 µM → ≥90% MMP inhibition in same assay
Often mediated via NF-κB/STAT3 downshift; some human biomarker data show reduced MMPs with green tea extracts.
VEGF/AngiogenesisPreclinical + limited human biomarker data
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: VEGF secretion (pg/mL) ↓ in tumor cells; HUVEC tube formation ↓ (%) at ≤10–20 µM
Anchor: 10 = Bevacizumab 10 µg/mL or SU5416 1 µM → near-complete tube inhibition
Anti-angiogenic effect is moderate compared with targeted anti-VEGF drugs.

Note: EGCG is generally safe but high doses may cause liver toxicity or interact with CYP450-metabolized drugs. Studied in: lung, breast, colorectal, prostate, and pancreatic cancers. Synergies include: Curcumin (Natural): Enhances apoptosis and PI3K inhibition in breast cancer. Resveratrol (Natural): Boosts DNMT inhibition and angiogenesis suppression in colorectal cancer. Quercetin (Natural): Synergizes to inhibit MMPs in prostate cancer.

References:

Essiac Tea

Early humanDetoxROS

Key Takeaway: A traditional herbal blend that may help reduce cancer-promoting inflammation and support liver and immune health, with some user-reported benefits.

Strength of Evidence: ⭐ Anecdotal — Mostly anecdotal data. Limited preclinical data and user reports; no strong clinical evidence for anti-cancer effects.

Mechanisms: Essiac Tea, a blend of burdock root, sheep sorrel, slippery elm, and Indian rhubarb root, exhibits antioxidant properties by scavenging reactive oxygen species (ROS), protecting DNA from damage. It may suppress NF-κB signaling through its herbal components, reducing inflammation-driven tumor growth. The herbs support detoxification via liver and immune modulation, potentially enhancing immune surveillance. Anecdotal and preclinical evidence suggests anti-inflammatory and detox effects, but mechanisms in cancer are not well-elucidated.

Detox/Carcinogen Binding & ClearanceAnecdote + limited preclinical
🔥 Potency2.5 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: Change in biomarkers of exposure (e.g., urinary mutagenicity or aflatoxin-DNA adducts) vs baseline
Anchor: 10 = Positive control like chlorophyllin or cholestyramine achieving ≥50–70% reduction in same biomarker
Direct, controlled Essiac-specific detox biomarker data are sparse; composition varies by brand.
Antioxidant/ROSIn vitro antioxidant + small human signals
🔥 Potency4.0 / 10
🧪 Confidence2 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability3 / 10
Endpoint: Intracellular ROS change by DCFDA (% vs control) in PBMCs or cell models; lipid peroxidation markers
Anchor: 10 = N-acetylcysteine 5 mM (or vitamin C 1 mM) → ≥80–90% ROS reduction in same assay
Consider timing away from ROS-dependent therapies (some chemo/radiation) to avoid blunting intended oxidative hits.

Note: Generally safe but may cause gastrointestinal upset or interact with diuretics. Studied in: general cancer care, with limited data on breast and prostate cancers. Synergies include: Curcumin (Natural): Enhances anti-inflammatory effects in breast cancer. Green Tea Extract (EGCG) (Natural): Boosts ROS scavenging and detoxification. Milk Thistle (Natural): Synergizes for liver support in cancer care.

References:

Fenbendazole

PreclinicalTubulinGlucosePDK-1

Key Takeaway: Dog dewormer shown to disrupt cancer cell structure and energy supply, stressing them into self-destruction, with promising lab results.

Strength of Evidence: ⭐⭐ Preclinical — Strong in vitro and animal data; human trials are limited and ongoing.

Mechanisms: Fenbendazole (FBZ), a benzimidazole anthelmintic, binds to tubulin, destabilizing microtubules and disrupting mitosis, leading to cell cycle arrest at G2/M phase. It downregulates GLUT-1, impairing glucose uptake, and inhibits PDK-1, shifting metabolism from glycolysis to oxidative phosphorylation, countering the Warburg effect. FBZ induces ROS-mediated apoptosis and modulates p53 pathways. Preclinical studies show selective toxicity to cancer cells in lung and hepatocellular models.

Tubulin/MicrotubulesIn vitro + in vivo (preclinical)
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability2 / 10
Endpoint: Tubulin polymerization inhibition or mitotic index (G2/M %) vs control at ≤1–5 µM FBZ
Anchor: 10 = Nocodazole 100 nM or Colchicine 1 µM → near-complete depolymerization/mitotic arrest
Human PK and safe exposure to achieve cytotoxic tumor levels are uncertain; bioavailability is low and hepatotoxicity has been reported.
Glucose Uptake (GLUT1)Preclinical
🔥 Potency4.5 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: 2-NBDG uptake reduction (% vs control) in tumor cells at ≤1–5 µM
Anchor: 10 = WZB117 10 µM or Phloretin 50 µM → ≥90% GLUT1-mediated uptake block
Downstream glycolysis impact depends on dose/exposure; combine score with PDK-1 metric for metabolic shift.
PDK-1 (metabolic switch)Preclinical
🔥 Potency3.5 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: PDK-1 activity or PDH E1α phosphorylation (Ser293) ↓ (% vs control) indicating PDH reactivation
Anchor: 10 = DCA 5 mM → strong PDH reactivation and glycolysis→OXPHOS shift in same model
Often indirect; effect sizes smaller than DCA at pharmacologic doses.

Note: Requires medical supervision due to potential hepatotoxicity and gastrointestinal side effects. Studied in: non-small cell lung, hepatocellular, and colorectal cancers. Synergies include: Metformin (Rx): Enhances glycolysis inhibition in lung cancer. Curcumin (Natural): Boosts ROS and apoptosis in hepatocellular cancer. Berberine (Natural): Synergizes to impair glucose uptake in colorectal cancer.

References:

Fisetin

PreclinicalSenolyticApoptosisPI3K

Key Takeaway: Fights cancer by clearing “zombie” senescent cells and pushing tumor cells toward apoptosis while dialing down PI3K/Akt growth signals; evidence is mainly preclinical.

Strength of Evidence: ⭐⭐ Preclinical — Strong in vitro and animal data; human trials are limited but ongoing.

Mechanisms: Fisetin, a flavonoid found in strawberries and apples, acts as a senolytic agent by selectively inducing apoptosis in senescent cells through activation of caspases-8 and -9, clearing 'zombie' cells that promote tumor growth. It inhibits PI3K/Akt/mTOR signaling, reducing proliferation, and upregulates pro-apoptotic proteins like Bax while downregulating anti-apoptotic Bcl-2. Fisetin also suppresses NF-κB and STAT3 pathways, limiting inflammation-driven cancer progression, and inhibits angiogenesis via VEGF downregulation. Preclinical studies demonstrate efficacy in breast, colon, and lung cancer models.

Senescent cell clearancePreclinical (senolysis in vitro/in vivo)
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability4 / 10
Endpoint: Loss of SA-β-gal⁺/p16INK4a⁺ senescent cells or viability ↓ (%) @24–72h in senescent HUVEC/IMR90 at ≤10–20 µM fisetin
Anchor: 10 = Dasatinib 100 nM + Quercetin 50 µM clearing ≥70% senescent cells in the same assay
Fisetin is among the stronger natural senolytics but generally weaker than D+Q in matched assays.
ApoptosisPreclinical (multiple tumor lines)
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Annexin V/PI–positive cells (% of total) @48h at ≤10–30 µM; caspase-3/7 activity fold-change
Anchor: 10 = Staurosporine 1 µM @24–48h yielding ≥70% apoptosis in the same line
Mitochondrial pathway (Bax↑/Bcl-2↓; caspase-3/9 cleavage) predominates.
PI3K/Akt/mTORPreclinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: p-Akt (Ser473) reduction (% vs control) @4–24h; downstream mTOR/S6K phosphorylation ↓
Anchor: 10 = MK-2206 1 µM or LY294002 10 µM causing ≥90% p-Akt reduction in the same model
Also modulates NF-κB/STAT3; those effects are not separately scored here.

Note: Generally safe at dietary doses but high doses may cause gastrointestinal upset or interact with CYP450-metabolized drugs. Studied in: breast, colorectal, lung, and prostate cancers. Synergies include: <ul><li><strong>Quercetin (Natural):</strong> Enhances senolytic effects and apoptosis in breast cancer. </li><li><strong>Dasatinib (Rx):</strong> Boosts clearance of senescent cells in lung cancer. </li><li><strong>Curcumin (Natural):</strong> Synergizes to inhibit PI3K/Akt in colorectal cancer.</li></ul>

References:

GcMAF

PreclinicalMacrophageAngio

Key Takeaway: Proposed macrophage activator with anti-angiogenic signals in early studies, but evidence is controversial and lacks robust clinical validation.

Strength of Evidence: ⭐ Controversial — Limited preclinical data; no strong clinical evidence, with ongoing debates.

Mechanisms: GcMAF (Gc protein-derived macrophage-activating factor) is proposed to activate macrophages by converting vitamin D-binding protein (Gc protein) into a form that stimulates macrophage phagocytosis and cytotoxicity against cancer cells. It may inhibit angiogenesis by suppressing endothelial cell proliferation and induce apoptosis in tumor cells. However, evidence is controversial, with limited preclinical data showing effects on cAMP formation and immune modulation, but no robust clinical validation due to regulatory issues and retracted studies.

Macrophage activationControversial/limited preclinical
🔥 Potency2.0 / 10
🧪 Confidence1 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability1 / 10
Endpoint: Phagocytosis index (% increase), CD86↑/HLA-DR↑ expression, or TNF-α/IL-12 secretion in human monocyte-derived macrophages ex vivo
Anchor: 10 = GM-CSF + IFN-γ priming achieving robust activation (e.g., ≥2× phagocytosis, strong CD86↑) in the same system
Literature quality is inconsistent; some reports retracted. Low confidence and translatability by design.
AngiogenesisControversial/limited preclinical
🔥 Potency1.5 / 10
🧪 Confidence1 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability1 / 10
Endpoint: Chick CAM assay or HUVEC tube formation: vessel area/tube length reduction (%)
Anchor: 10 = Bevacizumab (anti-VEGF) or SU5416 causing near-complete tube inhibition in the same assay
Signal present in select reports but not confirmed in rigorous clinical trials.

Note: Unapproved therapy with potential risks including injection site reactions; controversial due to lack of FDA approval and safety concerns. If considered, it should only be within properly regulated research settings. Potential complements (theoretical): Vitamin D repletion, lifestyle immune support.

References:

Gossypol (AT-101, Cottonseed Polyphenol)

Key Takeaway: A pan–Bcl-2 family (BH3-mimetic–like) natural product that can free up apoptosis machinery and modestly inhibit angiogenesis; the (-)-enantiomer AT-101 has human trial data with limited monotherapy efficacy but potential in combinations.

Strength of Evidence: ⭐⭐⭐ Moderate — Strong mechanistic and preclinical data; early-phase clinical trials with AT-101 show safety but limited efficacy as monotherapy. Potential in combination regimens still under study.

Mechanisms: Gossypol is a natural polyphenolic aldehyde from cottonseed that functions as a pan–Bcl-2 family inhibitor (‘BH3 mimetic’). By binding to anti-apoptotic proteins (Bcl-2, Bcl-xL, Mcl-1), it frees pro-apoptotic Bax/Bak, leading to mitochondrial outer-membrane permeabilization, cytochrome c release, and caspase activation. It also suppresses angiogenesis (VEGF ↓), disrupts NF-κB and PI3K/AKT signaling, and can radiosensitize or chemosensitize resistant tumors. The synthetic (-)-enantiomer AT-101 has been tested clinically in prostate, lung, head/neck, and lymphoid malignancies, but with modest responses.

Bcl-2 family (Bcl-2/Bcl-xL)Human phase I/II (AT-101) + strong preclinical
🔥 Potency4.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: Displacement of BH3 peptides from Bcl-2/Bcl-xL (Ki/IC50) or functional apoptosis rescue assays
Anchor: 10 = Venetoclax (Bcl-2-selective) or Navitoclax (Bcl-2/Bcl-xL) achieving near-complete target engagement in the same system
Gossypol binds broadly but with lower affinity than modern BH3 mimetics; off-target liabilities limit dosing.
Mcl-1Preclinical + limited human PD
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: Mcl-1 binding/inhibition (Ki/IC50) or dependence shift in BH3 profiling; MOMP induction
Anchor: 10 = S63845 or AZD5991 showing potent/ selective Mcl-1 inhibition in the same model
Partial Mcl-1 coverage may synergize with chemo or radiation.
ApoptosisPreclinical + early human biomarker signals
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Annexin V/PI % or cleaved caspase-3/PARP densitometry @24–48h in sensitive lines
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis in the same line
Mitochondrial (intrinsic) pathway via Bcl-2 family neutralization.
Angiogenesis/VEGFPreclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: VEGF secretion ↓ (pg/mL) and HUVEC tube formation ↓ (%)
Anchor: 10 = Bevacizumab or SU5416 achieving near-complete tube inhibition in the same assay
Anti-angiogenic effect is supportive rather than dominant vs BH3-mimetic action.

Note: Toxicities include gastrointestinal upset, fatigue, liver enzyme elevation, and historically, reversible infertility (studied as a male contraceptive). Clinical efficacy as AT-101 has been modest; not approved as a cancer drug. Use only under medical supervision or in trials. Combination strategies (with chemo/radiotherapy/targeted agents) are being explored.

References:

Honokiol

Key Takeaway: Magnolia bark compound that turns off STAT3/NF-κB growth switches, raises ROS to stress tumors, and pushes cells into apoptosis; promising in lab models, early human data limited.

Strength of Evidence: ⭐⭐ Preclinical — Strong in vitro and animal data; human trials are limited.

Mechanisms: Honokiol, a lignan from magnolia bark, inhibits STAT3 and NF-κB signaling, reducing inflammation-driven tumor growth and metastasis. It induces reactive oxygen species (ROS)-mediated apoptosis by disrupting mitochondrial function, upregulating caspases, and downregulating anti-apoptotic proteins like Bcl-2. Honokiol also suppresses PI3K/Akt/mTOR pathways, limiting cell proliferation, and inhibits angiogenesis via VEGF downregulation. Preclinical studies show efficacy in breast, lung, and colorectal cancer models, with potential to overcome drug resistance.

STAT3Preclinical (multiple tumor lines, xenografts)
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: p-STAT3 (Tyr705) ↓ (% vs control) and STAT3 reporter activity ↓ at ≤5–20 µM
Anchor: 10 = Stattic 5–10 µM (or equivalent selective STAT3 inhibitor) → ≥90% p-STAT3/reporter suppression in same assay
Downstream targets (Cyclin D1, Survivin) typically fall with STAT3 inhibition.
NF-κBPreclinical
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: NF-κB p65 nuclear translocation ↓ or κB-luciferase reporter ↓ (%)
Anchor: 10 = BMS-345541 (IKK inhibitor) 5 µM or Bay 11-7082 5 µM → ≥90% reporter inhibition
Often linked to IKK suppression and lowered IL-6/COX-2.
ROSPreclinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: DCFDA fluorescence fold-increase over control @≤24 h
Anchor: 10 = Menadione 25 µM (or H2O2 300 µM) producing high ROS signal in same cells
ROS rise precedes Δψm loss and caspase activation.
ApoptosisPreclinical
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Annexin V/PI % @24–48 h; cleaved caspase-3/PARP densitometry
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis in same line
Intrinsic (mitochondrial) pathway: Bax↑/Bcl-2↓, Δψm collapse.

Note: Generally safe but may cause gastrointestinal upset at high doses; interacts with CYP450-metabolized drugs. Studied in: breast, lung, colorectal, ovarian, and prostate cancers. Synergies include: <ul><li><strong>Rapamycin (Rx):</strong> Enhances apoptosis in breast cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts NF-κB inhibition and ROS in lung cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes to suppress STAT3 in colorectal cancer.</li></ul>

References:

Hydrogen Water

Early humanROS ScavengerOH

Key Takeaway: Molecular hydrogen–enriched water that scavenges hydroxyl radicals; early human data suggest it can reduce treatment side-effects and oxidative stress without clearly blunting anti-tumor therapy.

Strength of Evidence: ⭐⭐⭐ Moderate — Supported by preclinical studies and early human trials; larger RCTs needed.

Mechanisms: Hydrogen water selectively scavenges hydroxyl radicals (OH•), reducing oxidative stress while preserving beneficial ROS like hydrogen peroxide. It modulates redox balance, enhances immune function, and inhibits tumor growth by suppressing proliferation, inducing apoptosis, and reducing angiogenesis. Studies show it alleviates chemotherapy side effects, such as cisplatin-induced nephrotoxicity, by protecting normal cells without interfering with anti-cancer efficacy. Preclinical and early human trials demonstrate effects in colorectal, ovarian, and gastric cancers.

Global intracellular ROSEarly human biomarkers + preclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability6 / 10
Endpoint: DCFDA ROS signal ↓ (%) in PBMCs/tissues; urinary 8-OHdG ↓ (%) from baseline
Anchor: 10 = N-acetylcysteine 5 mM (ex vivo) or high-dose IV vitamin C producing ≥80–90% ROS signal reduction
Clinical benefit endpoints focus on symptom/toxicity reduction rather than direct tumor kill.
Hydroxyl radical (•OH)Preclinical + small trials
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity8.0 / 10
🎯 Translatability6 / 10
Endpoint: ESR spin-trapping (DMPO-OH) signal ↓ (%) in cell/biochemical systems
Anchor: 10 = Mannitol/thiourea scavenging at standard positive-control concentrations achieving near-complete •OH suppression
Selectivity for •OH helps spare signaling ROS (H2O2/NO).

Note: Generally safe with no known side effects at therapeutic doses; may interact with antioxidants. Consider timing around ROS-dependent therapies. Rare reports suggest context-dependent effects in highly oxidative tumors.

References:

Indole-3-Carbinol / DIM

Early humanEstrogen ModCYP1A1Apoptosis

Key Takeaway: Cruciferous compound pair that shifts estrogen metabolism toward 2-OH pathways, induces CYP1A1, and can trigger apoptosis; useful adjunct in hormone-sensitive settings with emerging human data.

Strength of Evidence: ⭐⭐⭐ Moderate — Supported by preclinical studies and early human trials; more RCTs needed for confirmation.

Mechanisms: Indole-3-carbinol (I3C) and its metabolite diindolylmethane (DIM) shift estrogen metabolism toward the protective 2-hydroxyestrone (2-OH) pathway by inducing CYP1A1 and other phase I enzymes, reducing estrogen-driven carcinogenesis. They inhibit NF-κB and AhR signaling, inducing apoptosis and cell cycle arrest in cancer cells. I3C/DIM also suppress hormone-sensitive tumor growth by downregulating ER-α and modulating androgen receptors. Preclinical and human studies show efficacy in breast, prostate, and cervical cancers.

Estrogen metabolism (2-OH:16α-OH ratio)Human biomarker change + preclinical
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: Urinary/serum 2-hydroxyestrone:16α-hydroxyestrone ratio ↑ (%) from baseline with oral I3C/DIM
Anchor: 10 = Strong AhR/CYP1A1 induction (e.g., β-naphthoflavone in vitro) or pharmacologic comparator doubling the ratio in the same model
Biomarker often improves at common supplemental doses of DIM/I3C.
CYP1A1 inductionPreclinical + human PD
🔥 Potency8.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability7 / 10
Endpoint: CYP1A1 mRNA/activity (EROD assay) fold-increase vs control after DIM/I3C
Anchor: 10 = β-naphthoflavone 10 µM (AhR agonist) producing maximal CYP1A1 induction in same system
AhR-mediated; downstream shift toward 2-OH metabolites.
ApoptosisPreclinical (hormone-sensitive tumor lines)
🔥 Potency4.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: Annexin V/PI % or caspase-3/7 activity ↑ at ≤10–50 µM
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis in same line
Also downregulates ER-α and modulates AR signaling depending on context.

Note: Generally safe but can alter hormone levels and drug metabolism; monitor when combined with endocrine therapy. Very high doses have shown liver effects in animals.

References:

Iodine (I₂ / I⁻)

Early humanPPARγApoptosisAngiogenesisEstrogen ModNIS ↕Thyroid Axis ⚠

Key Takeaway: Best studied as molecular iodine (I₂) with standard therapy—especially in breast cancer—where it may trigger PPARγ-driven apoptosis, temper angiogenesis, and improve chemo response in a small RCT. Because iodine strongly affects the thyroid and radioiodine treatments, use only with clinician oversight and avoid around planned I-131 therapy.

Strength of Evidence: ⭐⭐⭐ Moderate — Mechanistic and in vivo data plus a small randomized human study in breast cancer; benefits appear context- and dose-dependent. Separate rules apply in thyroid cancer/RAI care.

Mechanisms: Outside thyroid cancer care, I₂ can be converted to 6-iodolactone (6-IL), a PPARγ ligand that promotes mitochondrial apoptosis (Bax↑/Bcl-2↓, cytochrome-c release, caspase-3/7). Iodine can downshift VEGF/HIF-1α programs and modulate estrogen biology. A randomized, double-blind trial adding 5 mg/day I₂ to neoadjuvant chemo in breast cancer reported higher pCR rates and fewer AEs. High iodine competes with radioiodine uptake; avoid before I-123/I-131 imaging/therapy.

PPARγPreclinical + small human signals
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: PPARγ target gene induction (CD36, FABP4) and reporter activity after I₂/6-IL
Anchor: Comparable activation to 6-IL or rosiglitazone at equipotent levels
Driven by 6-iodolactone formation; cell-type dependent.
ApoptosisPreclinical + small human biomarker signals
🔥 Potency5.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability6 / 10
Endpoint: Cleaved caspase-3/7, TUNEL, Bax/Bcl-2 ratio in tumor/xenograft after I₂
Anchor: Reference cytotoxic (e.g., doxorubicin @ IC50) in same model
Magnitude modest vs standard cytotoxics.
VEGF/HIF-1α (Angiogenesis)Preclinical
🔥 Potency4.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability5 / 10
Endpoint: VEGF protein/mRNA and microvessel density (CD31⁺)
Anchor: Bevacizumab-like surrogate in same assay
Supportive effect; not dominant mechanism.
Estrogen ModulationPreclinical + limited human biomarkers
🔥 Potency4.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity3.0 / 10
🎯 Translatability5 / 10
Endpoint: ↑2-OH-estrone:16α-OH-estrone; ER-α activity readouts
Anchor: Shift magnitude vs standard endocrine therapy
Context/dose dependent signals.
NIS (Iodide Handling)Preclinical + human tissue expression
🔥 Potency3.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity2.0 / 10
🎯 Translatability4 / 10
Endpoint: NIS expression and radioiodide uptake in tumor tissue
Anchor: NIS-high reference tissue (thyroid) or NIS-upregulating controls
Breast NIS expression is variable; effects inconsistent.
Thyroid Axis (Safety)Established clinical physiology
🔥 Potency8.0 / 10
🧪 Confidence5 / 5
🛡 Selectivity2.0 / 10
🎯 Translatability9 / 10
Endpoint: TSH, free T4/T3 change; urinary iodine concentration
Anchor: Clinical thresholds used in thyroid practice
Wolff–Chaikoff/Jod-Basedow effects; avoid around radioiodine.

Note: Monitor TSH/free T4/T3 and urinary iodine if supplementing beyond diet. Avoid in active thyroiditis, uncontrolled hypo/hyperthyroidism, pregnancy unless prescribed, or if I-123/I-131 imaging/therapy is upcoming. Interacts with amiodarone, lithium; consider recent iodinated contrast. Clinical fit: Adjunct to standard therapy in selected breast cancer protocols (I₂ 5 mg/day studied) with thyroid monitoring. Avoid peri-RAI and in uncontrolled thyroid disease; coordinate with oncology/endocrinology.

References:

IP-6 + Myo-Inositol

Early humanNKChelatorAngio

Key Takeaway: Plant-derived combo that modestly chelates iron, boosts NK-cell activity, and downshifts angiogenesis/proliferation signals; small human studies suggest biomarker benefits and chemo support, but large oncology RCTs are scarce.

Strength of Evidence: ⭐⭐⭐ Moderate — Supported by preclinical studies and small human trials; larger RCTs ongoing.

Mechanisms: IP-6 (inositol hexaphosphate) and myo-inositol chelate iron, depriving cancer cells of essential metals for growth, while enhancing natural killer (NK) cell activity and immune surveillance. They inhibit angiogenesis by downregulating VEGF and suppress tumor proliferation through cell cycle arrest and apoptosis induction. The combination modulates PI3K/Akt and MAPK pathways, reducing metastasis. Human and animal studies show enhanced chemotherapy efficacy and reduced side effects in various cancers.

NK-cell cytotoxicitySmall human biomarker studies + preclinical
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: Ex vivo NK killing of K562 targets (% specific lysis) or CD107a degranulation ↑ from baseline
Anchor: 10 = IL-2 or lenalidomide producing ≥2× increase in NK cytotoxicity in the same assay
Increases IFN-γ/IL-12 in some studies; effect sizes modest–moderate.
Iron chelation↑ (chelating)Biochemical + cell models
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: Fe3+/Fe2+ binding (Kd/IC50) and ferritin iron mobilization; intracellular labile iron pool ↓
Anchor: 10 = Deferoxamine (100 µM) achieving near-complete LIP suppression in the same system
Chelation may contribute to anti-proliferative effects; avoid co-administration with iron therapy.
Angiogenesis (VEGF/Tube formation)Preclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: VEGF secretion ↓ (%) and HUVEC tube formation ↓ (%) vs control
Anchor: 10 = Sunitinib 50–100 nM abolishing tube formation in the same assay
Often accompanied by PI3K/Akt downshift and MMP-2/9 reductions.

Note: Generally safe but may cause gastrointestinal upset at high doses; interacts with iron supplements. Studied in: colorectal, prostate, breast, and lung cancers. Recent data suggest IP-6 may increase platelet aggregation and thrombosis risk in cancer patients (conflicting with anticoagulant claims). Synergies include: <ul><li><strong>Conventional Chemotherapy (Rx):</strong> Enhances anti-tumor effects and reduces metastases in colorectal cancer.</li><li><strong>Green Tea Extract (EGCG) (Natural):</strong> Synergizes to inhibit angiogenesis in some cancers.</li></ul>

References:

Itraconazole †Rx

Early humanHedgehogAngioCYP3A4

Key Takeaway: Antifungal with oncology repurposing signals: inhibits SMOGli (Hedgehog), exerts anti-angiogenic effects, and is a potent CYP3A4 inhibitor (PK booster). Phase I/II data show activity in select settings (e.g., prostate, NSCLC, BCC) though not yet standard of care.

Strength of Evidence: ⭐⭐⭐ Moderate — Supported by preclinical studies and phase I/II trials; larger RCTs needed.

Mechanisms: Itraconazole (ITZ), an antifungal, inhibits the Hedgehog pathway by blocking Smoothened (SMO) activation, reducing Gli transcription factors and tumor proliferation. It exerts anti-angiogenic effects by suppressing VEGF and endothelial cell migration, and inhibits CYP3A4, potentially enhancing chemotherapy efficacy. ITZ also induces autophagy, cell cycle arrest, and apoptosis while reversing multidrug resistance. Clinical trials show benefits in prostate, lung, and basal cell carcinoma.

Hedgehog (SMO→Gli)Preclinical + early clinical signal (e.g., BCC, prostate subsets)
🔥 Potency7.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability7 / 10
Endpoint: Gli1/2 mRNA ↓ (%) and Gli-luciferase reporter ↓ (%) in SMO-dependent models
Anchor: 10 = Vismodegib/Sonidegib at clinically relevant concentrations achieving near-complete Gli suppression
Acts at SMO but by a distinct binding mechanism compared with vismodegib.
AngiogenesisPreclinical + biomarker changes in patients
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: HUVEC tube formation/migration ↓ (%); VEGFR2 phosphorylation ↓
Anchor: 10 = Sunitinib 50–100 nM abolishing tube formation in the same assay
Clinical imaging/biomarker studies show vascular changes with ITZ.
CYP3A4 (drug–drug interaction)↓ (inhibition)Established clinical PK effect
🔥 Potency9.0 / 10
🧪 Confidence5 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability10 / 10
Endpoint: Human liver microsome CYP3A4 activity ↓ (midazolam 1′-hydroxylation rate) and clinical ↑AUC of CYP3A4 substrates
Anchor: 10 = Ketoconazole 1 µM (prototypic strong CYP3A4 inhibitor) producing near-complete activity suppression
Explains part of the ‘PK-boosting’ phenomenon when combined with some chemotherapies.

Note: Requires medical supervision due to hepatotoxicity/QT/cardiac risks; major CYP3A4 and P-gp interactions. Studied in: prostate, lung, basal cell, and breast cancers. Synergies include: <ul><li><strong>Docetaxel (Rx):</strong> Enhanced responses in metastatic prostate cancer subsets.</li><li><strong>Paclitaxel (Rx):</strong> Synergistic antitumor activity in colorectal and ovarian cancers.</li><li><strong>Pemetrexed (Rx):</strong> Second-line therapy in NSCLC.</li><li><strong>5-FU (Rx):</strong> Inhibits gastric cancer growth.</li><li><strong>Hydroxychloroquine (Rx):</strong> Synergistic via lysosomal function.</li></ul> Safety: Monitor liver function tests; risk of heart failure/edema; contraindicated with certain drugs like dofetilide, quinidine.

References:

Ivermectin †Rx

PreclinicalWntMitoApoptosis

Key Takeaway: Antiparasitic that inhibits Wnt/β-catenin signaling, disrupts mitochondrial potential/respiration, and triggers ROS-mediated apoptosis; compelling preclinical data with emerging human signals, but clinical efficacy in oncology remains to be proven.

Strength of Evidence: ⭐⭐ Preclinical — Strong in vitro and animal data; human trials limited but ongoing.

Mechanisms: Ivermectin (IVM), an antiparasitic drug, inhibits the Wnt/TCF signaling pathway, reducing β-catenin nuclear translocation and downregulating genes involved in tumor growth and metastasis. It collapses mitochondrial membrane potential, inducing mitochondrial dysfunction, oxidative damage, and ROS-mediated apoptosis via caspase activation. IVM also modulates Akt/mTOR and MAPK pathways, leading to cell cycle arrest and enhanced chemotherapy sensitivity. Preclinical studies show selective toxicity to cancer cells in renal, breast, and lung models.

Wnt/β-catenin–TCFPreclinical (reporters, target genes, xenografts)
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability3 / 10
Endpoint: TOPFlash/TCF-luciferase reporter ↓ (%); nuclear β-catenin ↓; Axin2/c-Myc transcript ↓
Anchor: 10 = XAV939 5–10 µM or LGK974 100 nM yielding ≥90% reporter suppression in the same assay
Downstream proliferation/EMT genes typically fall with Wnt blockade.
Mitochondrial membrane potential (Δψm)Preclinical
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: JC-1 red/green ratio ↓ or TMRE signal ↓ (%) at ≤24 h; OCR (Seahorse) ↓
Anchor: 10 = CCCP 10 µM causing near-complete depolarization in the same cells
Δψm loss precedes caspase-3/7 activation and ROS surge.
ApoptosisPreclinical
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Annexin V/PI %, cleaved caspase-3/7 and PARP ↑ at pharmacologic concentrations
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis at 24 h in the same model
Often ROS-dependent; NAC can blunt the effect in vitro.

Note: Requires medical supervision due to potential neurotoxicity at high doses; interacts with CYP450-metabolized drugs. Studied in: renal, breast, lung, and esophageal cancers. Synergies include: <ul><li><strong>Doxorubicin (Rx):</strong> Enhances apoptosis in breast cancer.</li></ul>

References:

Kaempferol

PreclinicalVEGFHIF-1αApoptosis

Key Takeaway: Dietary flavonoid that lowers HIF-1α/VEGF signaling (anti-angiogenic), nudges PI3K/Akt and ERK down, and triggers caspase-mediated apoptosis with G2/M arrest in multiple tumor models. Promising as an adjunct; human oncology trials remain limited.

Strength of Evidence: ⭐⭐ Preclinical — Robust cell/animal data; human trials limited.

Mechanisms: Kaempferol suppresses VEGF and HIF-1α under hypoxia, inhibiting angiogenesis; modulates Bcl-2/Bax with caspase-3/9 activation to induce apoptosis; arrests cells at G2/M via JNK/p38; and inhibits PI3K/Akt and ERK signaling to reduce proliferation and migration. Preclinical efficacy spans colon, breast, lung, and ovarian cancers.

Angiogenesis (VEGF/HIF-1α)Preclinical (hypoxia models, HUVEC assays)
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: VEGF secretion ↓ (%) and HUVEC tube formation ↓ (%) under 1–2% O₂
Anchor: 10 = Sunitinib 50–100 nM abolishing tube formation in the same assay
Often accompanied by HIF-1α protein↓ and p-Akt↓.
ApoptosisPreclinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Annexin V/PI %, caspase-3/9 cleavage, Bax/Bcl-2 ratio
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis at 24 h in the same model
JNK/p38 activation commonly precedes caspase signaling.
PI3K/AktPreclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: p-Akt (Ser473) ↓ (%) and downstream p-mTOR/p70S6K ↓
Anchor: 10 = MK-2206 (1–5 µM) producing near-complete p-Akt suppression
Contributes to G2/M arrest and reduced migration.

Note: Generally safe at dietary intake; concentrated supplements may cause GI upset. Watch for additive effects with other anti-angiogenic or PI3K-pathway agents. Synergies: <ul><li><strong>Cisplatin (Rx):</strong> Enhanced apoptosis in ovarian models.</li><li><strong>Quercetin (Natural):</strong> Additive cytostasis/apoptosis in breast/colon models.</li><li><strong>Curcumin (Natural):</strong> Boosts anti-angiogenic effects in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for apoptosis induction in breast cancer.</li></ul> Safety: Rare allergic reactions; potential estrogenic activity at high doses—caution in hormone-sensitive cancers.

References:

Kelp Flakes

PreclinicalIodineFucoidanApoptosis

Key Takeaway: Whole-food seaweed providing iodine and fucoidan: lab data suggest iodine-triggered apoptosis in some breast cancer models and fucoidan-mediated anti-angiogenic/anti-metastatic and immune-support effects. Use food amounts; high iodine can disturb thyroid function.

Strength of Evidence: ⭐⭐ Preclinical — Strong in vitro/animal signals; human trials limited.

Mechanisms: Kelp supplies iodine (and molecular iodine species) that can induce caspase-dependent apoptosis and alter estrogen signaling in breast models; fucoidan, a sulfated polysaccharide, inhibits proliferation, EMT, invasion (MMP-2/9↓), and angiogenesis (VEGF↓), and can activate NK cells and ROS-mediated tumor cell death. Evidence spans breast, colon, lung, and prostate models.

Apoptosis (iodine-induced)Preclinical (breast cancer lines, iodine/iodolipid exposure)
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: Annexin V/PI %, caspase-3 cleavage after molecular iodine exposure
Anchor: 10 = Staurosporine 1 µM causing ≥70% apoptosis at 24 h
Effects vary with ER status and iodine species/dose.
Angiogenesis/EMT (fucoidan)Preclinical
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: VEGF secretion ↓ (%), HUVEC tube formation ↓ (%), MMP-2/9 activity ↓
Anchor: 10 = Sunitinib 50–100 nM abolishing tube formation
Often accompanied by E-cadherin↑ and vimentin↓.
NK-cell activity (fucoidan)Preclinical ± limited human biomarker
🔥 Potency4.0 / 10
🧪 Confidence2 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Ex vivo NK cytotoxicity (K562 lysis %) or CD107a degranulation ↑
Anchor: 10 = IL-2 producing ≥2× increase in NK cytotoxicity
Magnitude modest; dietary matrix may influence bioactivity.

Note: Keep iodine intake within recommended limits; avoid in hyperthyroidism or with iodine-containing meds unless supervised. ER-α interactions are context-dependent. Synergy: <ul><li><strong>Tamoxifen (Rx):</strong> With iodine/fucoidan shows additive apoptosis in breast models.</li><li><strong>Cisplatin (Rx):</strong> Fucoidan enhances anti-metastatic effects in lung models.</li><li><strong>Curcumin (Natural):</strong> Boosts NK activity and apoptosis in colon cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for VEGF inhibition in prostate models.</li></ul> Safety: Monitor thyroid function; potential for iodine overload (e.g., acne, metallic taste).

References:

Lactoferrin

Early humanIronNKp53

Key Takeaway: Iron-binding glycoprotein that can starve tumors of labile iron, nudge p53-mediated apoptosis, and modestly raise NK-cell activity. Early human data (prevention/QoL, immune markers) exist; oncology outcomes are limited and context-dependent.

Strength of Evidence: ⭐⭐⭐ Moderate — Preclinical + early clinical immune/biomarker data; mixed context signals and limited oncology outcome trials.

Mechanisms: Lactoferrin chelates Fe³⁺/Fe²⁺ and downshifts the labile iron pool (LIP), limiting Fenton chemistry and DNA synthesis; modulates p53/p21 and caspase cascades to induce cell-cycle arrest/apoptosis; increases NK cytotoxicity and Th1 signaling; dampens NF-κB–linked inflammation; and shows anti-angiogenic effects via VEGF suppression.

Iron availability (tumor LIP)Preclinical + small human biomarker studies
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: Labile iron pool (calcein-quench) ↓ %, transferrin saturation/NTBI
Anchor: 10 = Deferasirox or Deferoxamine producing ≥80% LIP reduction in the same model
Effect magnitude depends on baseline iron status and dosing form (bovine, recombinant).
NK-cell activityHuman biomarker + preclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability6 / 10
Endpoint: Ex vivo NK cytotoxicity (K562 lysis %) or CD107a+ degranulation
Anchor: 10 = IL-2/IL-15 activation roughly doubling NK cytotoxicity in the same assay
Typical increases ~10–40% over baseline in small trials.
p53 pathway / apoptosisPreclinical
🔥 Potency3.5 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: p53 stabilization (Ser15 phosphorylation), p21↑, caspase-3/7 activity
Anchor: 10 = Nutlin-3 (10 µM) causing robust p53 accumulation and apoptosis in the same line
Context-dependent; strongest in models with intact p53.

Note: Generally safe (common in dairy/infant formulas); occasional GI upset. Iron interplay: avoid taking simultaneously with iron supplements (separate by ≥2–3 h). Evidence is heterogeneous and cancer-type specific; pro-growth signals have been reported in some contexts—use clinically with monitoring. Synergies: <ul><li><strong>Cisplatin (Rx):</strong> Enhanced apoptosis via iron depletion in lung models.</li><li><strong>Curcumin (Natural):</strong> Boosts NK activity and p53 stabilization in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Additive anti-angiogenic effects in breast cancer.</li><li><strong>Quercetin (Natural):</strong> Synergizes for inflammation dampening in prostate models.</li></ul> Safety: Rare hypersensitivity; monitor ferritin/iron levels in long-term use.

References:

Low-Dose Naltrexone (LDN)

Early humanTregOpioid ModEndorphin

Key Takeaway: Nightly 1–4.5 mg naltrexone transiently blocks opioid receptors, provoking a rebound in endogenous endorphins and modulating the OGF–OGFr axis; small studies suggest lowered Tregs and possible antiproliferative/immune effects as an adjunct. Larger oncology RCTs are still sparse.

Strength of Evidence: ⭐⭐⭐ Moderate — Preclinical + small clinical studies suggest immune/endocrine modulation and adjunct potential; confirmatory oncology RCTs are limited.

Mechanisms: Short opioid-receptor blockade → rebound β-endorphin/met-enkephalin → activation of opioid growth factor (OGF) pathway, cell-cycle modulation (p16/p21), and immune effects (Treg downshift, Th1 tilt). Preclinical/early clinical signals in colorectal, neuroblastoma, breast, and others; may enhance chemo sensitivity and QoL.

Regulatory T cells (Tregs)Early human + preclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: CD4+CD25+FoxP3+ fraction (%) among CD4 T cells; suppressive function assays
Anchor: 10 = Low-dose cyclophosphamide producing ~40–50% Treg depletion/function loss
Magnitude varies; best framed as immunomodulation rather than depletion.
Endorphin/OGF axisHuman physiologic + preclinical oncology
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: Serum β-endorphin/met-enkephalin (pg/mL) rise 3–6 h post-dose; OGFr signaling markers
Anchor: 10 = Exogenous enkephalin/OGF therapy driving near-maximal OGFr activation in the same system
Night dosing optimizes rebound; avoid concurrent opioid agonists.
Proliferation/apoptosis (OGF–OGFr)↓ proliferation / ↑ apoptosisPreclinical + small clinical signals
🔥 Potency4.0 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: Ki-67 ↓ (%), cell-cycle arrest (p16/p21), caspase-3/7 ↑
Anchor: 10 = Cytotoxic chemo causing ≥80% Ki-67 reduction in responders
Greatest promise appears as an adjunct within multimodal regimens.

Note: Rx and supervision required. Common: vivid dreams/insomnia initially; generally well-tolerated. Time dosing at night to leverage rebound endorphins. Consider interactions with concurrent opioids (can blunt analgesia). Synergies: <ul><li><strong>Chemotherapy (Rx):</strong> Enhanced response in colorectal cancer.</li><li><strong>Cannabidiol (Natural):</strong> Increases anticancer efficacy in some models.</li><li><strong>Curcumin (Natural):</strong> Boosts immune modulation in breast cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for OGF pathway activation in neuroblastoma.</li></ul> Safety: Rare hepatotoxicity; monitor LFTs in long-term use; avoid in acute opioid withdrawal.

References:

Maitake D-Fraction / Lentinan

Clinicalβ-Glucan PRRCTLNK

Key Takeaway: β-Glucan mushroom extracts activate innate/adaptive immunity (NK and CTL) and can improve outcomes alongside chemotherapy—best signals in GI cancers.

Strength of Evidence: ⭐⭐⭐⭐ Strong — Multiple trials/meta-analyses (especially lentinan) show adjunct survival/immune benefits in GI cancers.

Mechanisms: Maitake D-fraction and lentinan (β-1,3/1,6-glucans) engage Dectin-1/CR3 → Syk/NF-κB signaling, maturing APCs and enhancing Th1 cytokines. This boosts NK cytotoxicity and CD8⁺ CTL function, curbing tumor growth and improving chemo responses. Meta-analyses in gastric/colorectal cancer show OS benefits with lentinan adjunct therapy.

β-Glucan PRR (Dectin-1/CR3)Human biomarker + clinical adjunct trials
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: Monocyte/DC activation (CD80/86↑), IL-12/IFN-γ↑; opsonized β-glucan → CR3 priming
Anchor: 10 = Potent TLR agonist producing maximal APC activation
Oral bioactivity depends on particle size/processing; IV lentinan used in trials.
Cytotoxic T-lymphocytes (CD8⁺)Preclinical + human signals
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: CD8⁺ IFN-γ ELISPOT↑; tumor-infiltrating CD8⁺ density↑; granzyme B/perforin↑
Anchor: 10 = Checkpoint inhibitor level CTL reinvigoration
Often mediated by improved antigen presentation.
NK-cell cytotoxicityHuman biomarker + clinical adjunct trials
🔥 Potency5.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability7 / 10
Endpoint: Ex vivo K562 lysis % or CD107a+ degranulation ↑ from baseline
Anchor: 10 = IL-2/IL-15 producing ~2× NK activity in same assay
NK gains correlate with symptom and QoL improvements in some cohorts.

Note: Generally safe; rare allergy. Use caution in active autoimmunity or with strong immunosuppressants. Synergies: <ul><li><strong>5-FU (Rx):</strong> Improves OS in gastric cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts NK activity in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for immune modulation in liver cancer.</li><li><strong>EGCG (Natural):</strong> Enhances APC maturation in lung models.</li></ul> Safety: Mild GI upset possible; monitor for hypersensitivity in mushroom-allergic patients.

References:

Mebendazole †Rx

Key Takeaway: Anthelmintic repurposed for oncology: Potent tubulin disruptor with anti-angiogenic and pro-apoptotic actions; strong preclinical efficacy and emerging phase I/II signals, particularly in brain and gynecologic cancers.

Strength of Evidence: ⭐⭐⭐ Moderate — Robust preclinical data; phase I/II trials show safety and signals in glioma/ovarian; larger RCTs ongoing.

Mechanisms: Mebendazole (MBZ) inhibits microtubule polymerization by binding β-tubulin, disrupting mitosis and inducing G2/M arrest; suppresses VEGF/HIF-1α signaling for anti-angiogenic effects; activates intrinsic apoptosis via caspase-3/9, Bax upregulation, and p53 stabilization; additional targets include Hedgehog, NF-κB, and kinase pathways; penetrates BBB effectively; preclinical and early clinical data in glioma, ovarian, colorectal, and other solid tumors.

Tubulin/MicrotubulesPreclinical + phase I PK/PD
🔥 Potency8.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability7 / 10
Endpoint: Microtubule polymerization ↓ (%), G2/M arrest (flow cytometry %), mitotic index ↓
Anchor: 10 = Paclitaxel 10 nM achieving near-complete spindle disruption
Nanosensitivity; synergizes with taxanes.
Angiogenesis (VEGF/HIF-1α)Preclinical (HUVEC/tumor models)
🔥 Potency6.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: VEGF secretion ↓ (ELISA %), tube formation ↓ (%), HIF-1α stabilization ↓
Anchor: 10 = Bevacizumab neutralizing >90% VEGF in same assay
Hypoxia-relevant; contributes to anti-metastatic effects.
ApoptosisPreclinical + early clinical
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: Annexin V/PI %, caspase-3/9 cleavage, Bax/Bcl-2 ratio ↑
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis at 24h
p53-dependent in many lines; enhanced by combo therapies.

Note: Requires medical supervision; potential hepatotoxicity—monitor LFTs monthly. Generally well-tolerated at anti-parasitic doses; higher oncology regimens (e.g., 100-500 mg BID) may cause GI upset or neutropenia. Synergies: <ul><li><strong>Temozolomide (Rx):</strong> Enhanced survival in glioma trials.</li><li><strong>Cisplatin (Rx):</strong> Overcomes resistance in ovarian cancer.</li><li><strong>Docetaxel (Rx):</strong> Synergistic microtubule effects in prostate models.</li><li><strong>Curcumin (Natural):</strong> Boosts apoptosis in colorectal cancer.</li></ul> Safety: Contraindicated in hepatic impairment; avoid with strong CYP3A4 inducers.

References:

Melatonin

Key Takeaway: Circadian hormone that activates SIRT1/3, downshifts mTOR, and promotes apoptosis; repeatedly improves chemo-tolerance and sometimes response rates in clinical studies.

Strength of Evidence: ⭐⭐⭐⭐ Strong — Multiple clinical trials support toxicity reduction and adjunct benefits; mechanistic depth is substantial.

Mechanisms: Melatonin engages MT1/MT2 and mitochondrial targets to activate SIRT1/3, improving DNA repair and metabolic efficiency. It inhibits PI3K/Akt/mTOR signaling, enhances p53/p21, and increases caspase-mediated apoptosis. As an adjunct, it reduces chemo/radiotherapy toxicity (myelosuppression, mucositis, neurotoxicity) and may improve response in specific settings.

Sirtuins (SIRT1/3)Human adjunct trials + preclinical PD
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: SIRT1/3 activity↑; downstream targets (PGC-1α, FOXO) deacetylation
Anchor: 10 = Potent SIRT activator achieving maximal deacetylation in same assay
Mitochondrial protection contributes to lower treatment toxicity.
mTOR/PI3K-AktPreclinical + human biomarker signals
🔥 Potency5.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: p-Akt/p-mTOR/p-S6 ↓ (%) in tumor/immune cells; proliferation markers ↓
Anchor: 10 = Everolimus/rapamycin near-complete mTOR blockade
Pathway shift aligns with improved chemo response in some trials.
Apoptosis (p53/caspases)Preclinical + clinical adjunct
🔥 Potency5.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: Annexin V/PI %, cleaved caspase-3/7, Bax/Bcl-2 ratio
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis
Often synergizes with chemo/radiation.

Note: Generally safe; may cause drowsiness or vivid dreams. Time at night (e.g., 10–40 mg in oncology studies; dosing varies by protocol). Synergies: <ul><li><strong>Doxorubicin (Rx):</strong> Enhances apoptosis in breast cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts SIRT1 activation in lung cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for mTOR inhibition in colorectal cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies antioxidant protection in prostate models.</li></ul> Safety: Rare headaches; caution in autoimmune conditions or with sedatives.

References:

Metformin †Rx

ClinicalAMPKmTORCSC

Key Takeaway: Metabolic reprogrammer that activates AMPK, suppresses mTOR/insulin-IGF signaling, and targets cancer stem-cell phenotypes; broad human data (esp. in diabetics) show risk-reduction and outcome signals.

Strength of Evidence: ⭐⭐⭐⭐ Strong — Extensive epidemiology + multiple trials/meta-analyses; oncology-specific RCTs mixed but promising in subsets.

Mechanisms: Metformin lowers hepatic gluconeogenesis and circulating insulin/IGF-1, activating AMPK (↑AMP:ATP) and inhibiting mTORC1 (via TSC2/Rheb). Tumor-intrinsic effects include decreased protein synthesis, cell-cycle arrest, and CSC downshift (ALDH⁺/CD44⁺↓). It can sensitize tumors to chemo/radiation and may reduce recurrence risk in select cohorts.

AMPK activationEstablished clinical pharmacology + oncology biomarkers
🔥 Potency7.0 / 10
🧪 Confidence5 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability9 / 10
Endpoint: p-AMPK (Thr172)↑; downstream ACC phosphorylation↑
Anchor: 10 = Direct AMPK agonist achieving maximal activation
Systemic insulin/IGF lowering augments tumor-intrinsic AMPK effects.
mTORC1 signalingPreclinical + human signals
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability8 / 10
Endpoint: p-S6K/p-4EBP1 ↓ (%) in tumor/immune cells
Anchor: 10 = Rapalog producing near-complete pathway suppression
Magnitude depends on insulin/IGF tone and dose.
Cancer stem-cell traits (CSC)Preclinical + observational clinical
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: ALDH⁺/CD44⁺ fractions ↓; sphere formation ↓; EMT markers (ZEB1/Snail) ↓
Anchor: 10 = Targeted CSC inhibitor abolishing sphere formation
Best as part of multi-agent metabolic/CSC strategy.

Note: Rx only. GI upset common initially; rare lactic acidosis risk increases with severe renal/hepatic impairment. Consider vitamin B12 monitoring long-term. Synergies: <ul><li><strong>Doxorubicin (Rx):</strong> Enhances efficacy in breast cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts AMPK activation in lung cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for mTOR inhibition in colorectal cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies CSC targeting in prostate models.</li></ul> Safety: Contraindicated in eGFR <30; dose adjust for moderate impairment.

References:

Methylene Blue

PreclinicalRedox CyclingETC Bypass¹O₂

Key Takeaway: Ancient redox dye that can shuttle electrons to bypass ETC defects and, when light/ultrasound-activated, generates singlet oxygen to kill tumor cells; compelling preclinical data with emerging clinical use in PDT/SDT.

Strength of Evidence: ⭐⭐ Preclinical — Robust cell/animal data; growing early-phase clinical experience in PDT/SDT settings.

Mechanisms: MB accepts/donates electrons (MB⁺/leucomethylene blue), supporting mitochondrial respiration when Complex I/III are impaired. As a photo/sono-sensitizer, excited MB transfers energy to O₂ to form ¹O₂, driving lipid/protein/DNA oxidation → apoptosis/necrosis. It modulates ROS and can synergize with other oxidant therapies.

Mitochondrial ETC (bypass/complex I–III)Preclinical + limited human PD
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: OCR (Seahorse) rescue and Δψm restoration with MB in ETC-impaired cells
Anchor: 10 = Direct electron donor fully restoring ETC flux
Effect is dose/context dependent; excessive redox cycling can increase ROS.
Redox cycling / ROS modulationPreclinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5 / 10
Endpoint: DCFDA ROS signal fold-change; GSH:GSSG shift; caspase activation downstream
Anchor: 10 = Menadione/H₂O₂ control producing maximal ROS
Low doses may be antioxidant; in PDT/SDT contexts, pro-oxidant effects dominate.
Singlet oxygen (¹O₂) generation (PDT/SDT)Preclinical + early clinical PDT
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: ¹O₂ phosphorescence/ESR signals; tumor necrosis/apoptosis post-illumination
Anchor: 10 = Benchmark PDT sensitizer achieving near-maximal ¹O₂ yield
Effect requires light/ultrasound at appropriate wavelength/energy; tissue penetration limits apply.

Note: Medical supervision advised at higher doses/PDT. May discolor urine/sclera; avoid with strong serotonergic drugs at high doses (theoretical risk). Synergies: <ul><li><strong>Cisplatin (Rx):</strong> Enhances ROS-mediated apoptosis in ovarian models.</li><li><strong>Curcumin (Natural):</strong> Boosts PDT efficacy in breast cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for mitochondrial protection in lung cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies singlet oxygen yield in colorectal PDT.</li></ul> Safety: Monitor for methemoglobinemia at IV doses >7 mg/kg; contraindicated in G6PD deficiency.

References:

Mistletoe (VAE) †Rx

Early humanLectin (RIP-II)QoLCytokinesNKApoptosis

Key Takeaway: Injectable Viscum album extracts can trigger lectin-mediated tumor apoptosis and modulate immunity (NK/CTL, IL-12/TNF-α). Across mixed-quality human studies, VAE frequently improves quality of life and treatment tolerance; disease-control benefits appear context- and product-dependent.

Strength of Evidence: ⭐⭐⭐ Moderate — Consistent QoL signals with adjunct use; survival/response benefits vary and depend on extract and setting.

Mechanisms: VAE contains lectins (RIP-II) and viscotoxins. Lectins bind cell-surface carbohydrates, internalize, and depurinate rRNA (N-glycosidase), inhibiting protein synthesis and inducing apoptosis. Pattern-recognition signaling (e.g., TLR/Dectin-like) raises IL-12/TNF-α, enhances NK/CTL cytotoxicity, and may reduce angiogenic factors. Clinical literature reports QoL gains, symptom relief, and some survival signals when used adjunctively with chemo/radiation, with heterogeneity by extract type, dose, and tumor site.

Lectin/RIP-II cytotoxicityPreclinical + small clinical signals
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5 / 10
Endpoint: rRNA depurination/protein synthesis block; Annexin V/PI and caspase-3/7 activation in treated cells/tumors
Anchor: 10 = Potent RIP achieving near-complete protein synthesis inhibition in same assay
Potency varies by extract (e.g., ABNOBAviscum, Iscador) and glycoform content.
Cytokine milieu (IL-12, TNF-α, IL-6)Human biomarker + preclinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: Serum/ PBMC cytokines; Th1 skewing and DC activation markers
Anchor: 10 = Strong innate agonist producing maximal IL-12/TNF-α
Transient pyrogenic responses common early in titration.
NK-cell cytotoxicityHuman biomarker + adjunct trials
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: Ex vivo K562 lysis %, CD107a degranulation, granzyme B/perforin upregulation
Anchor: 10 = IL-2/IL-15 doubling NK activity in same assay
NK increases correlate with symptom/QoL gains in some cohorts.
Quality of life / symptom burdenMultiple human trials/observational studies
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability8 / 10
Endpoint: EORTC QLQ-C30 global health, fatigue/pain subscales; chemo dose intensity maintained
Anchor: 10 = Intervention producing large, consistent QLQ-C30 improvements across domains
Effect sizes modest–moderate; survival impact inconsistent.

Note: Rx-only; typically subcutaneous (slow up-titration). Monitor for local reactions, fever, hypotension, or rare allergy. Use caution in autoimmune disease and with immunotherapies until coordinated. Products are not interchangeable—stick to a single standardized brand. Synergies: <ul><li><strong>Gemcitabine (Rx):</strong> Improved tolerance in pancreatic cancer.</li><li><strong>Cisplatin (Rx):</strong> Enhanced NK response in lung models.</li><li><strong>Curcumin (Natural):</strong> Boosts cytokine modulation in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for apoptosis in breast cancer.</li></ul> Safety: Flu-like symptoms common (manage with dose adjustment); rare anaphylaxis.

References:

Modified Citrus Pectin

Early humanGalectin-3AdhesionMetastasisImmune

Key Takeaway: Soluble, low–molecular-weight pectin binds galectin-3, disrupting tumor cell adhesion, aggregation, and microenvironmental signaling; small human studies (notably in prostate cancer) show biomarker improvements and disease-stabilization signals.

Strength of Evidence: ⭐⭐⭐ Moderate — Promising Gal-3 blockade with small human studies; larger RCTs ongoing.

Mechanisms: MCP blocks galectin-3 carbohydrate recognition domains, preventing lattice formation that supports cell–cell/ECM adhesion, angiogenesis, and immune evasion. This can reduce circulating tumor cell aggregation, tumor emboli, and metastatic colonization. Additional effects include macrophage polarization shifts and microbiome interactions.

Galectin-3 binding/neutralizationPreclinical + small human PD
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: Gal-3 CRD occupancy; inhibition of Gal-3–mediated cell aggregation/adhesion
Anchor: 10 = High-affinity Gal-3 inhibitor abolishing lattice formation
Low–MW/low–DE MCP improves bioavailability and Gal-3 binding.
Adhesion/Metastatic seedingPreclinical + early clinical signals
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: CTC aggregation ↓; endothelial adhesion/transendothelial migration ↓; metastasis counts in vivo
Anchor: 10 = Potent anti-adhesive abolishing CTC emboli formation
Human data include PSA kinetics and time-to-progression in small prostate cohorts.
Immune microenvironmentPreclinical + limited human biomarker
🔥 Potency3.5 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability5 / 10
Endpoint: Macrophage polarization (M1/M2) and T-cell activation markers
Anchor: 10 = Strong immunomodulator producing large M1 shift
Immune effects secondary to Gal-3 blockade; clinical significance still emerging.

Note: Generally well tolerated; mild GI effects possible. Separate dosing from certain oral meds to avoid binding in the gut. Use standardized, low–MW/low–DE MCP products for better absorption. Synergies: <ul><li><strong>Docetaxel (Rx):</strong> Reduced PSA progression in prostate cancer.</li><li><strong>Cisplatin (Rx):</strong> Enhanced anti-metastatic effects in ovarian models.</li><li><strong>Curcumin (Natural):</strong> Boosts galectin-3 blockade in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for adhesion inhibition in breast cancer.</li></ul> Safety: Rare bloating; monitor PSA/biomarkers in prostate use.

References:

Nattokinase

PreclinicalFibrinThrombolysisPlateletMetastasis

Key Takeaway: Clot-dissolving protease from natto that degrades fibrin and enhances plasmin activity, potentially disrupting platelet–fibrin cloaking that aids metastasis. Oncology data are mostly preclinical; bleeding risk is the major clinical concern.

Strength of Evidence: ⭐⭐ Moderate — Strong fibrinolytic/antithrombotic evidence; oncology benefits remain largely preclinical and inferential.

Mechanisms: Nattokinase (subtilisin-like serine protease) directly cleaves fibrin and increases endogenous fibrinolysis (plasmin generation), while modestly inhibiting platelet aggregation and improving microcirculation. By dismantling fibrin/platelet scaffolds, it might reduce tumor cell adhesion and vascular trapping; however, controlled human oncology trials are scarce.

Fibrin degradation / fibrinolysisHuman cardiovascular + preclinical oncology
🔥 Potency7.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability8 / 10
Endpoint: Clot lysis time↓; D-dimer/ plasmin–antiplasmin complexes↑; plasma fibrinogen↓
Anchor: 10 = Therapeutic tPA achieving rapid complete lysis
Systemic lysis less predictable than tPA; oral bioactivity debated but supported by some PK data.
Platelet aggregationPreclinical + limited human
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: ADP/AA-induced platelet aggregation %↓ ex vivo
Anchor: 10 = Potent antiplatelet abolishing aggregation
Additive bleeding risk with aspirin/DOACs/warfarin.
Metastatic support (fibrin/platelet cloaking)Preclinical oncology
🔥 Potency3.5 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: CTC survival/adhesion ↓; experimental lung colony counts ↓
Anchor: 10 = Targeted anti-metastatic abolishing seeding
Mechanistic rationale plausible; robust human cancer data lacking.
Safety: bleeding riskClinical pharmacology/ case reports
🔥 Potency7.0 / 10
🧪 Confidence5 / 5
🛡 Selectivity3.0 / 10
🎯 Translatability10 / 10
Endpoint: Bleeding events, INR/platelet function changes when combined with anticoagulants
Anchor: 10 = Full-dose anticoagulation level bleeding risk
Discontinue before surgery; avoid with coagulopathy or intracranial lesions.

Note: Avoid with anticoagulants/antiplatelets or bleeding disorders; stop before invasive procedures. Standardize units (FU) across brands; quality varies. Consider safer, trial-backed antithrombotic strategies first in active cancer. Synergies: <ul><li><strong>Cisplatin (Rx):</strong> Reduced thrombosis in ovarian models.</li><li><strong>Curcumin (Natural):</strong> Boosts fibrinolytic effects in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for platelet inhibition in prostate cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies anti-metastatic in lung models.</li></ul> Safety: Monitor for bruising/bleeding; contraindicated in hemorrhagic stroke history.

References:

Niraparib †Rx

ClinicalPARPHRD Synthetic Lethality

Key Takeaway: Oral PARP1/2 inhibitor with clinically proven maintenance and treatment benefits in HRD/BRCA-altered tumors—most robust in ovarian cancer—by blocking PARP repair and trapping PARP on DNA to exploit synthetic lethality.

Strength of Evidence: ⭐⭐⭐⭐⭐ Strong — FDA-approved with multiple phase III trials and meta-analyses demonstrating PFS benefit, strongest in HRD/BRCA settings.

Mechanisms: Niraparib inhibits PARP catalytic activity and traps PARP-DNA complexes, stalling replication forks and converting single-strand lesions into double-strand breaks. HRD/BRCA-deficient cells cannot repair these DSBs by homologous recombination → apoptosis. Phase III trials show significant PFS gains in ovarian cancer (including all-comer maintenance with strongest effect in HRD-positive subgroups). Combination strategies (e.g., with bevacizumab) and expansion to other HRR-deficient tumors are supported by growing evidence.

PARP1/2 catalytic activityPhase III trials + pharmacodynamic biomarkers
🔥 Potency7.5 / 10
🧪 Confidence5 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability10 / 10
Endpoint: PARylation levels ↓ in tumor/PBMCs at therapeutic doses
Anchor: 10 = Complete catalytic blockade with maximal PAR depletion
On-target effect across tumor and normal tissues; clinical efficacy depends on HRD context.
PARP–DNA complex trappingPreclinical ranking + clinical correlation
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability9 / 10
Endpoint: Chromatin-bound PARP ↑; replication fork stalling (comet assay, RAD51)
Anchor: 10 = Highest-trapping PARPi in class in matched assays
Trapping potency is a key driver of cytotoxicity and myelosuppression.
DNA damage/HRD synthetic lethality↑ (selective kill)Phase III efficacy in HRD/BRCA tumors
🔥 Potency8.0 / 10
🧪 Confidence5 / 5
🛡 Selectivity8.0 / 10
🎯 Translatability10 / 10
Endpoint: γH2AX foci ↑; reduced RAD51 foci formation; PFS benefit in HRD+
Anchor: 10 = HRD-perfect biomarker selection yielding maximal clinical benefit
Greatest benefit in BRCA1/2-mutated or HRD-positive disease; activity extends to some HRR gene alterations.

Note: Monitor CBCs (thrombocytopenia, anemia), blood pressure (hypertension), and fatigue. Individualized dosing by baseline weight/platelets reduces hematologic AEs. Drug–drug interactions are fewer than some PARPis but review co-meds. Synergies: <ul><li><strong>Bevacizumab (Rx):</strong> Extended PFS in ovarian maintenance.</li><li><strong>Pembrolizumab (Rx):</strong> Enhanced response in HRD-positive endometrial cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts PARP inhibition in preclinical ovarian models.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for HRD exploitation in breast cancer.</li></ul> Safety: MDS/AML risk ~1-2%; contraception required (teratogenic).

References:

Nivolumab †Rx

ClinicalPD-1T-Cell CytotoxicityirAE

Key Takeaway: Anti–PD-1 checkpoint inhibitor that restores exhausted T cells and delivers survival gains across multiple cancers (melanoma, NSCLC, RCC, HNSCC, others); requires vigilant monitoring for immune-related toxicities.

Strength of Evidence: ⭐⭐⭐⭐⭐ Strong — FDA-approved across many cancers with consistent OS/PFS benefits in phase III trials.

Mechanisms: By binding PD-1 on T cells, nivolumab prevents engagement with PD-L1/PD-L2, reversing T-cell exhaustion and boosting cytotoxic activity and memory responses. Tumor control stems from renewed CD8⁺ infiltration and effector function. Durable response ‘tails’ on survival curves occur in subsets with immunogenic tumors or high neoantigen burden.

PD-1 signalingMultiple phase III trials across tumor types
🔥 Potency9.0 / 10
🧪 Confidence5 / 5
🛡 Selectivity8.0 / 10
🎯 Translatability10 / 10
Endpoint: Receptor occupancy; PD-1/PD-L1 axis blockade with T-cell reinvigoration gene signature
Anchor: 10 = Complete functional PD-1 blockade in vivo
Pharmacodynamic exhaustion-reversal signatures predict responders.
CD8⁺ T-cell cytotoxicity/infiltrationClinical biomarker + translational studies
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability9 / 10
Endpoint: TIL density↑, granzyme B/perforin↑, IFN-γ signatures, clonal expansion
Anchor: 10 = Maximal CTL activation seen with combinatorial ICI
Tumor mutational burden/PD-L1 and inflamed TME enrich for benefit but are imperfect.
Safety: immune-related AEsBroad clinical experience
🔥 Potency6.0 / 10
🧪 Confidence5 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability10 / 10
Endpoint: Incidence/severity of grade ≥3 irAEs; steroid response rates
Anchor: 10 = High-toxicity ICI combinations
Early recognition and guideline-based management preserve efficacy and safety.

Note: Monitor for irAEs: dermatitis, colitis/diarrhea, hepatitis (LFTs), pneumonitis, thyroiditis/hypo-hyperthyroidism, hypophysitis. Use standardized steroid algorithms; hold or discontinue per grade. Synergies: <ul><li><strong>Ipilimumab (Rx):</strong> OS benefit in melanoma and RCC.</li><li><strong>Chemotherapy (Rx):</strong> Enhanced response in NSCLC.</li><li><strong>Curcumin (Natural):</strong> Reduces irAEs in preclinical models.</li><li><strong>Resveratrol (Natural):</strong> Boosts T-cell function in lung cancer.</li></ul> Safety: Infusion reactions rare; endocrine irAEs may be permanent—replace hormones as needed.

References:

Omega-3 (EPA/DHA)

ClinicalInflammationCachexiaChemo Tolerance

Key Takeaway: Marine omega-3s generate specialized pro-resolving mediators (SPMs) that quell inflammation, help preserve weight/lean mass, and can improve treatment tolerance and select outcomes in cancer cachexia.

Strength of Evidence: ⭐⭐⭐⭐ Strong — Multiple trials and meta-analyses support anti-inflammatory and anti-cachexia roles; effect sizes moderate and protocol-dependent.

Mechanisms: EPA/DHA incorporate into membranes, shifting eicosanoid profiles away from arachidonic acid derivatives and spawning resolvins/protectins/maresins. This reduces NF-κB/COX-2 signaling and cytokines (IL-6/TNF). In cachexia, omega-3s help stabilize weight and lean mass, temper anorexia/inflammation, and may enhance chemo efficacy via membrane fluidity and lipid raft effects.

Systemic inflammation (NF-κB/COX-2, cytokines)Human RCTs/meta-analyses
🔥 Potency5.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8 / 10
Endpoint: CRP/IL-6/TNF↓; NF-κB activity↓; SPM levels↑
Anchor: 10 = Potent anti-inflammatory drug achieving near-complete suppression
Benefits depend on dose, baseline omega-3 index, and product quality.
Cancer cachexia (weight/lean mass)↓ (loss)Human RCTs/mixed meta-analyses
🔥 Potency5.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8 / 10
Endpoint: Body weight/FFM stabilization; appetite/QoL scales
Anchor: 10 = Anabolic/anti-catabolic drug fully reversing cachexia
Greatest effects when combined with nutrition/exercise and adequate protein.
Chemotherapy tolerance/efficacyPreclinical + clinical signals
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: Dose intensity maintained; GI toxicity/fatigue↓; response rate signals in subsets
Anchor: 10 = Standard supportive care achieving maximal tolerance
Membrane/lipid-raft modulation and SPMs may enhance chemo sensitivity in some settings.

Note: Generally safe; may increase bleeding tendency at high doses or with anticoagulants. Use purified, oxidatively stable products; consider 2–4 g/day EPA+DHA in cachexia protocols used in trials. Synergies: <ul><li><strong>Cisplatin (Rx):</strong> Improves tolerance in lung cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts anti-inflammatory effects in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for cachexia management in pancreatic models.</li><li><strong>Quercetin (Natural):</strong> Amplifies SPM production in breast cancer.</li></ul> Safety: GI upset rare; fishy aftertaste minimized with enteric coating; monitor lipids if hypertriglyceridemia.

References:

PEA (Palmitoylethanolamide)

Early humanPPARNeuro-InflammationNeuropathic Pain

Key Takeaway: Endogenous fatty-acid amide that activates PPAR-α to calm mast cells/microglia and reduce neuropathic pain; promising adjunct for chemotherapy-induced peripheral neuropathy (CIPN) with supportive human data.

Strength of Evidence: ⭐⭐⭐ Moderate — Human neuropathy studies and mechanistic rationale support use; oncology-specific RCTs for CIPN are emerging.

Mechanisms: PEA binds/activates PPAR-α and engages the ‘ALIA’ mechanism (Autacoid Local Injury Antagonism) to inhibit mast-cell degranulation and microglial activation. Downstream, it lowers pro-inflammatory cytokines and neuronal hyperexcitability, easing allodynia/hyperalgesia. Studies show benefit in diabetic neuropathy and emerging signals in CIPN; ultra-micronized forms may improve bioavailability.

PPAR-α activationPreclinical + human PD
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: PPAR-α target genes (CPT1A, ACOX1)↑; inflammatory gene expression↓
Anchor: 10 = Potent PPAR-α agonist reaching maximal transcriptional response
PPAR-α–dependent and –independent (ALIA) mechanisms both contribute.
Neuroinflammation (mast cells/microglia)Preclinical + small clinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: MC degranulation↓; Iba1⁺ microglia activation↓; IL-6/TNF/NGF↓
Anchor: 10 = Strong glial inhibitor abolishing activation
Ultra-micronized formulations show greater clinical signals.
Neuropathic pain/CIPN symptomsHuman observational/RCT signals (non-oncology > oncology)
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: Pain intensity (VAS/NRS)↓; neuropathy scales (CIPN20)↓
Anchor: 10 = Gold-standard neuropathic analgesic response
Best as part of multimodal CIPN management (exercise, ALA, Mg, dose adjustments).

Note: Generally well tolerated; occasional GI upset/somnolence. Consider ultra-micronized PEA (um-PEA) 600–1200 mg/day used in studies; can be paired with standard CIPN measures. Synergies: <ul><li><strong>Duloxetine (Rx):</strong> Enhanced pain relief in CIPN trials.</li><li><strong>Curcumin (Natural):</strong> Boosts PPAR-α activation in neuroinflammation models.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for microglial calming in diabetic neuropathy.</li><li><strong>Quercetin (Natural):</strong> Amplifies mast-cell stabilization in allergic contexts.</li></ul> Safety: Rare headaches; monitor in bipolar disorder (theoretical mood effects).

References:

Phosphatidylcholine

Early humanMembraneBile FlowHepatotoxicity

Key Takeaway: Replenishes cell and mitochondrial membranes, improves bile composition/flow, and helps protect the liver from chemo-related steatohepatitis and transaminitis. Human data suggest hepatoprotective and anti-inflammatory effects that can support treatment tolerance.

Strength of Evidence: ⭐⭐⭐ Moderate — Convergent preclinical and small clinical data for hepatoprotection; larger oncology trials needed.

Mechanisms: As a major phospholipid, phosphatidylcholine (PC) restores membrane bilayers, stabilizes lipid rafts, and improves mitochondrial function under oxidative stress. In the liver, PC enriches biliary phospholipids, buffering bile-acid cytotoxicity, enhancing bile flow, and reducing cholestatic injury. PC modulates choline metabolism (SAMe/PC cycle), downshifts hepatic inflammation and stellate-cell activation, and can limit fibrosis progression.

Membrane repair/mitochondrial stabilityPreclinical + small human signals
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: Membrane phospholipid composition normalization; mitochondrial respiration/Δψm preservation under oxidative stress
Anchor: 10 = Pharmacologic membrane stabilizer fully normalizing mitochondrial metrics
Supports hepatocyte resilience during chemotherapy.
Bile acid cytotoxicity buffering/flow↑ (protection/flow)Preclinical + clinical physiology
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: Bile phospholipid:BA ratio↑; cholestatic biomarkers (ALP/GGT)↓
Anchor: 10 = Reference bile-acid therapy fully correcting cholestasis
Higher biliary PC reduces BA-induced epithelial injury.
Chemo-related hepatotoxicitySmall clinical studies
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: ALT/AST peaks↓; steatohepatitis scores↓; fewer dose reductions
Anchor: 10 = Gold-standard hepatoprotectant abolishing enzyme flares
Signals seen with polyenylphosphatidylcholine in steatohepatitis models and small oncology cohorts.

Note: Generally well tolerated; occasional GI upset. Consider choline intake/trimethylamine-N-oxide (TMAO) context in vascular disease. Use standardized PC (e.g., polyenylphosphatidylcholine) when possible in hepatoprotection protocols. Synergies: <ul><li><strong>Sorafenib (Rx):</strong> Reduced hepatotoxicity in HCC trials.</li><li><strong>Curcumin (Natural):</strong> Boosts membrane stabilization in pancreatic models.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for anti-fibrotic effects in liver cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies bile flow in cholestatic settings.</li></ul> Safety: Rare fishy odor; monitor liver enzymes in long-term use.

References:

Piperine (from Black Pepper, Piper nigrum)

Key Takeaway: Powerful bioavailability enhancer that inhibits intestinal P-gp and CYP3A4, markedly raising exposure to co-administered agents (e.g., curcumin). Preclinical oncology data show pro-apoptotic, anti-invasive, and chemosensitizing effects, but there are no human cancer-outcome trials and drug–drug interactions can be significant.

Strength of Evidence: ⭐⭐ Preclinical — Robust mechanistic and PK data as a bioenhancer; no human cancer-outcome trials; high interaction risk.

Mechanisms: Acute piperine exposure inhibits ABCB1 (P-gp) and CYP3A4, increasing oral AUC/Cmax of co-agents. In tumor models, piperine raises ROS, activates caspases, suppresses NF-κB/STAT3/PI3K-Akt, and lowers MMP-2/9 and VEGF, reducing invasion/angiogenesis. It can sensitize tumors (e.g., docetaxel synergy) partly via PK and efflux modulation. Repeated exposure may engage PXR with complex effects on CYP expression.

P-gp (ABCB1) effluxHuman ex vivo + clinical PK
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: Transport inhibition in Caco-2/MDCK; ↑AUC of P-gp substrates in humans
Anchor: 10 = Potent clinical P-gp inhibitor abolishing efflux
Explains large increases in co-administered nutraceutical/drug exposure.
CYP3A4 metabolism↓ (inhibition, ± induction with chronic PXR)In vitro + human PK
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability7 / 10
Endpoint: Midazolam 1′-hydroxylation↓; substrate AUC↑
Anchor: 10 = Strong clinical CYP3A4 inhibitor
Time- and dose-dependent; chronic use may induce via PXR.
Apoptosis (caspase/JNK)Preclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Annexin V/PI %, cleaved caspase-3/9; JNK activation
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis
Magnitude model-dependent; clinical anti-tumor proof lacking.
Invasion/angiogenesis (MMP-2/9, VEGF)Preclinical (in vitro/in vivo)
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Gelatin zymography↓; HUVEC tube formation↓
Anchor: 10 = Reference anti-angiogenic abolishing tubes
Partly secondary to NF-κB/STAT3 suppression.

Note: High interaction risk—avoid with chemotherapy, anticoagulants, or narrow-therapeutic-index drugs unless supervised. Human evidence supports PK enhancement (e.g., curcumin ↑ ~2000%), not cancer control. Synergies: <ul><li><strong>Docetaxel (Rx):</strong> Enhanced efficacy in prostate models.</li><li><strong>Curcumin (Natural):</strong> Dramatic bioavailability boost (~2000%).</li><li><strong>Resveratrol (Natural):</strong> Synergizes for NF-κB inhibition in colorectal cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies anti-angiogenic effects in breast models.</li></ul> Safety: GI irritation at high doses; monitor liver enzymes with chronic use.

References:

Piperlongumine (Piplartine)

PreclinicalROSGSTP1ApoptosisChemo-Sensitizer

Key Takeaway: Tumor-biased pro-oxidant that inhibits GSTP1 and perturbs thiol systems to push cancer cells past their oxidative limit, triggering apoptosis and chemosensitization in models. Promising preclinical breadth, but no therapeutic human trials to date.

Strength of Evidence: ⭐⭐ Preclinical — Broad mechanistic and xenograft activity; lacks human therapeutic trials; early landmark paper retracted, but independent confirmation exists.

Mechanisms: Piperlongumine elevates ROS and impairs antioxidant defenses by inhibiting GSTP1 and affecting thioredoxin-linked pathways, activating JNK/MAPK, cleaving caspases, and inducing apoptosis (and sometimes autophagy). It suppresses NF-κB/STAT3/PI3K-Akt signaling, reduces migration/invasion, and synergizes with platinums and other cytotoxics in xenografts. Note: the influential 2011 report was retracted in 2018, though independent studies continue to show ROS-mediated anticancer activity.

Reactive oxygen species (ROS)Preclinical (cells/xenografts)
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability3 / 10
Endpoint: DCFDA/ESR ROS signal↑; reversible with NAC
Anchor: 10 = Strong pro-oxidant producing near-maximal ROS
Tumor-biased due to higher basal oxidative stress.
GSTP1 (detox enzyme)Biochemical + cellular
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability3 / 10
Endpoint: GSTP1 activity/occupancy↓; downstream GSH conjugation↓
Anchor: 10 = Potent GSTP1 inhibitor abolishing activity
Direct binding reported; contributes to redox collapse.
Apoptosis (JNK/MAPK → caspases)Preclinical
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability3 / 10
Endpoint: Annexin V/PI %; cleaved caspase-3/9; PARP cleavage
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis
Often blocked by NAC or GSTP1 rescue.
Chemosensitization (platinums, etc.)Preclinical in vivo/in vitro
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability3 / 10
Endpoint: Synergy indices<1; tumor growth delay↑ in xenografts
Anchor: 10 = Established clinical chemosensitizer
Mechanism via redox stress and survival-pathway suppression.

Note: Experimental agent with no human efficacy data yet. Distinct from piperine. Potential for off-target oxidative toxicity; avoid during pregnancy. Consider only in research contexts. Synergies: <ul><li><strong>Cisplatin (Rx):</strong> Enhanced efficacy in cervical cancer models.</li><li><strong>Auranofin (Rx):</strong> Dual GSTP1/TrxR inhibition in breast cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts ROS/apoptosis in colorectal models.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for NF-κB suppression in head & neck cancer.</li></ul> Safety: Hepatotoxicity possible; monitor LFTs in preclinical dosing.

References:

Quercetin

Early humanSenolyticEGFRHSP27

Key Takeaway: Polyphenol with senolytic activity (especially paired with dasatinib) that can dampen EGFR/HSP27 stress-survival signaling, nudge apoptosis/autophagy, and modestly chelate iron; early human signals with stronger preclinical support.

Strength of Evidence: ⭐⭐⭐ Moderate — Preclinical breadth with early human signals; oncology RCTs are limited.

Mechanisms: Quercetin (QUE) contributes to senescent-cell clearance in D+Q regimens by tipping mitochondrial apoptosis (caspase-8/9/3 activation) and suppressing SASP outputs. It weakly inhibits EGFR kinase signaling and downregulates HSP27/HSPB1, reducing pro-survival chaperoning and metastasis traits. As a metal chelator, QUE lowers labile iron and Fenton chemistry, limiting DNA/lipid oxidation. Additional actions include NF-κB/STAT3 downshift, autophagy induction, and MMP/VEGF reduction.

Senescent cell clearance (D+Q)Preclinical + early human biomarker
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability5 / 10
Endpoint: Loss of SA-β-gal⁺/p16INK4a⁺ cells; Annexin V/PI in senescent models with QUE±Dasatinib
Anchor: 10 = Dasatinib 100 nM + Quercetin 50 µM clearing ≥70% senescent cells in matched assays
QUE alone is milder; pairing with dasatinib enhances senolysis.
EGFR signalingPreclinical
🔥 Potency4.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: p-EGFR (Tyr1068) and downstream p-ERK/p-AKT ↓ (% vs control)
Anchor: 10 = Clinically active EGFR TKI achieving near-complete pathway blockade
Magnitude is modest relative to TKIs; acts as a supportive modulator.
HSP27/HSPB1 (chaperone)Preclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: HSP27 expression/phosphorylation ↓; apoptotic priming ↑
Anchor: 10 = Direct HSP27 inhibitor abolishing client chaperoning
Lower HSP27 may reduce invasion/chemo-resistance.

Note: Generally safe at dietary intakes; high-dose supplements may cause GI/renal strain or drug interactions (OAT transporters/CYPs). Consider pulsed dosing for senolysis with medical supervision; separate from metal/iron dosing. Synergies: <ul><li><strong>Dasatinib (Rx):</strong> Enhanced senescent clearance in oral cancer.</li><li><strong>Cisplatin (Rx):</strong> Boosts apoptosis in breast cancer.</li><li><strong>Curcumin (Natural):</strong> Additive cytostasis in breast/colon models.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for iron chelation in colon cancer.</li></ul> Safety: Kidney strain at >1 g/day; estrogenic at high doses—caution in ER+ cancers.

References:

Reishi Ganoderma

Early humanβ-GlucanTRAILNK

Key Takeaway: Ganoderma lucidum polysaccharides and triterpenes activate innate/adaptive immunity (NK/CTL) and sensitize tumor cells to TRAIL-mediated apoptosis; human studies show immune/QoL gains with adjunct use.

Strength of Evidence: ⭐⭐⭐ Moderate — Human immune/QoL data and solid preclinical mechanisms; large oncology RCTs limited.

Mechanisms: β-Glucans engage Dectin-1/CR3 → Syk/NF-κB and APC maturation, boosting Th1 cytokines and NK/CTL activity. Ganoderic acids upregulate DR4/DR5 and inhibit NF-κB, enhancing TRAIL-induced apoptosis. Additional effects include anti-angiogenesis (VEGF↓), MMP-2/9↓, and microbiome–immune crosstalk that can improve therapy tolerance.

β-Glucan PRR (Dectin-1/CR3)Human biomarker + clinical adjunct
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: APC activation (CD80/86↑), IL-12/IFN-γ↑; CR3 priming
Anchor: 10 = Potent innate agonist producing maximal APC activation
Oral bioactivity depends on particle size/process; hot-water extracts common.
TRAIL-mediated apoptosis (DR4/DR5)Preclinical + translational
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: Death receptor expression↑; caspase-8/3 cleavage with TRAIL
Anchor: 10 = Potent TRAIL-sensitizer restoring apoptosis in resistant cells
Triterpenes (ganoderic acids) are primary contributors.
NK-cell cytotoxicityHuman biomarker + clinical
🔥 Potency5.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability7 / 10
Endpoint: Ex vivo NK lysis %, CD107a degranulation; IFN-γ response
Anchor: 10 = IL-2/IL-15 doubling NK activity
Correlates with QoL/immune symptom improvements in small trials.

Note: Generally safe; rare allergy or GI upset. Standardize to β-glucan/triterpene content; products vary widely. Use as an adjunct, not a monotherapy. Synergies: <ul><li><strong>Chemotherapy (Rx):</strong> Enhances apoptosis in lung cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts NK activity in breast cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for TRAIL sensitization in colorectal cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies anti-angiogenic effects in prostate models.</li></ul> Safety: Mild digestive issues; monitor in autoimmune conditions.

References:

Resveratrol

Early humanSirtuinNF-κBApoptosis

Key Takeaway: Pleiotropic polyphenol that activates SIRT1, suppresses NF-κB–driven inflammation, and promotes apoptosis; adjunct potential with mixed but encouraging human signals.

Strength of Evidence: ⭐⭐⭐ Moderate — Early human studies plus extensive preclinical data; definitive oncology RCTs remain limited.

Mechanisms: Resveratrol (RSV) activates SIRT1→FOXO/PGC-1α deacetylation, enhancing DNA repair/mitochondrial efficiency and reducing senescence signaling. It inhibits NF-κB (IKK/p65), lowering COX-2/IL-6/TNF and angiogenic VEGF. RSV primes intrinsic apoptosis (Bax↑/Bcl-2↓, caspase-3/9) and can modulate ER/AR signaling in hormone-responsive tumors. Synergistic effects with chemo/radiation are reported preclinically and in small trials.

Sirtuins (SIRT1)Preclinical + human biomarker
🔥 Potency5.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: SIRT1 activity/target deacetylation (FOXO/PGC-1α)↑; mitochondrial biogenesis markers
Anchor: 10 = Potent SIRT activator achieving maximal deacetylation
May reduce therapy-related fatigue via mitochondrial effects.
NF-κB/Inflammatory signalingPreclinical + small trials
🔥 Potency5.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: p65 nuclear translocation/κB-reporter↓; CRP/IL-6/TNF↓
Anchor: 10 = Strong anti-inflammatory agent abolishing NF-κB activity
Angiogenesis (VEGF) often drops in parallel.
Apoptosis (intrinsic)Preclinical + early clinical signals
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: Annexin V/PI %, caspase-3/9 cleavage, Bax/Bcl-2 ratio
Anchor: 10 = Staurosporine 1 µM → ≥70% apoptosis
Pro-apoptotic effects are context- and dose-dependent.

Note: Well tolerated at dietary doses; high-dose supplements may cause GI upset or interact with anticoagulants/CYP substrates. Bioavailability is low—consider micronized/trans-resveratrol formulations if used. Synergies: <ul><li><strong>Doxorubicin (Rx):</strong> Enhances efficacy in breast cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts NF-κB inhibition in colorectal cancer.</li><li><strong>Quercetin (Natural):</strong> Synergizes for apoptosis in prostate cancer.</li><li><strong>EGCG (Natural):</strong> Amplifies SIRT1 activation in lung models.</li></ul> Safety: Rare estrogenic effects; monitor in hormone-sensitive cancers.

References:

Selenium (Stand-alone)

ClinicalGPxTrxRp53 Apoptosis

Key Takeaway: Cofactor for selenoenzymes (GPx, TrxR) that buffers redox stress; certain selenium metabolites (e.g., methylselenol) can push p53-mediated apoptosis. Clinical signals are context-dependent—benefit is more likely when correcting deficiency; indiscriminate high-dose use can be harmful.

Strength of Evidence: ⭐⭐⭐ Moderate — Mixed clinical data: correcting deficiency shows the clearest benefit; prevention trials in selenium-replete populations are neutral/negative. Prefer status-guided, form-specific use.

Mechanisms: Selenium incorporates into glutathione peroxidases (GPx) and thioredoxin reductases (TrxR), lowering peroxides and stabilizing redox signaling. Pro-apoptotic selenium metabolites (methylselenol) enhance p53 transactivation, DNA damage responses, and caspase cleavage, selectively stressing tumor cells with impaired antioxidant reserve. In adjunct settings, selenium may reduce chemo/radiotoxicity and modulate immunity; prevention data are mixed and strongly moderated by baseline selenium status and chemical form.

Glutathione peroxidases (GPx) activityHuman PD + RCTs (mixed by baseline status)
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8 / 10
Endpoint: RBC/plasma GPx activity↑; lipid peroxidation markers↓
Anchor: 10 = Maximal GPx induction with complete peroxide buffering
Greatest upshift when correcting deficiency; ceiling effects in replete states.
Thioredoxin reductase (TrxR)Preclinical + human PD
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability7 / 10
Endpoint: TrxR activity↑; downstream redox-sensitive transcription normalized
Anchor: 10 = Pharmacologic TrxR upregulation with robust redox reset
Form and dose influence TrxR kinetics.
p53-mediated apoptosisPreclinical + limited human biomarker
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: p53 target activation, caspase-3/9 cleavage, Annexin V/PI↑
Anchor: 10 = Potent DNA-damaging agent causing ≥70% apoptosis
Pro-apoptotic effects linked to methylselenol generation; tumor-context dependent.

Note: Narrow therapeutic window—optimize (not exceed) status (e.g., serum/plasma selenium, selenoprotein P). Chronic excess → selenosis (hair/nail changes, GI, neuropathy). Forms differ (selenomethionine ≠ methylselenocysteine); keep daily supplemental doses modest unless deficiency is documented. Avoid routine high-dose co-supplementation with vitamin E for prostate cancer prevention (large RCT signals harm in replete men). Potential adjacencies: may mitigate platinum-induced toxicities and support redox balance during therapy under supervision. Synergies: <ul><li><strong>Cisplatin (Rx):</strong> Reduces nephrotoxicity in lung cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts apoptosis in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for redox protection in prostate cancer.</li><li><strong>Vitamin E (Natural):</strong> Antioxidant combo in lung cancer, but caution in prostate (SELECT trial harm).</li></ul> Safety: Test baseline status; upper limit 400 mcg/day; monitor for selenosis signs.

References:

Sodium Butyrate

PreclinicalHDACTumor-Suppressor ↑

Key Takeaway: SCFA and class I/IIa HDAC inhibitor that re-expresses tumor suppressors and can trigger cell-cycle arrest/apoptosis—strong preclinical data; human oncology trials are limited. Clinically, leverage diet/tributyrin/colon-targeted delivery rather than high-dose sodium butyrate.

Strength of Evidence: ⭐⭐ Preclinical — Strong mechanistic/animal data with limited oncology trials; prioritize diet- and colon-targeted delivery.

Mechanisms: Butyrate inhibits HDACs, increasing histone H3/H4 acetylation and transcription of p21, p27, and pro-apoptotic genes. It modulates NF-κB and fuels colonocytes, improving barrier function and dampening inflammation. Epigenetic reprogramming reduces proliferation, induces differentiation, and sensitizes to 5-FU and other agents in colorectal and breast models.

Histone deacetylases (HDAC) activityPreclinical + early human biomarker
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: H3/H4 acetylation↑; HDAC enzyme activity↓; chromatin accessibility↑
Anchor: 10 = Clinical-grade HDAC inhibitor with near-complete target blockade
Local colonic exposure > systemic; delivery route matters.
Tumor-suppressor gene expressionPreclinical
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: p21/p27 upregulation; cell-cycle arrest (G1); apoptosis markers↑
Anchor: 10 = Targeted epigenetic drug restoring suppressors in vivo
Synergizes with 5-FU and polyphenols in CRC models.
Colonic epithelial metabolism/inflammation↑ (support) / ↓ (inflammation)Human physiology + preclinical oncology
🔥 Potency5.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability8 / 10
Endpoint: Barrier markers↑, NF-κB activity↓, SCFA levels↑
Anchor: 10 = Robust clinical anti-inflammatory reversal
Dietary fiber and tributyrin reliably raise colonic butyrate.

Note: Generally safe via diet (fermentable fiber/resistant starch) and well-tolerated tributyrin; high oral sodium butyrate can cause GI upset and sodium load. Consider microbiome-forward strategies (fiber + probiotics that raise luminal butyrate). Potential synergies: 5-FU (CRC; apoptosis ↑), curcumin (HDAC & NF-κB co-modulation), resveratrol (complementary epigenetic re-expression).

References:

Soursop (Graviola)

PreclinicalComplex-IROSEMT

Key Takeaway: Acetogenins inhibit mitochondrial Complex I, elevate ROS, and can downshift EMT/invasion—yielding selective cytotoxicity in models. Human therapeutic data are lacking, and high exposure has neurotoxicity signals; treat as experimental.

Strength of Evidence: ⭐⭐ Preclinical — Compelling mechanisms but no therapeutic human trials; safety concerns at high exposure.

Mechanisms: Annona acetogenins (e.g., annonacin) bind NADH dehydrogenase (Complex I), collapsing mitochondrial ATP production and increasing ROScaspase activation and apoptosis/necrosis. Parallel suppression of PI3K/Akt/NF-κB and EMT transcription factors (Snail/ZEB1) reduces migration and invasion. Combination studies suggest additivity with platinums and polyphenols via oxidative and metabolic stress.

Mitochondrial Complex I (NADH dehydrogenase)Preclinical (cells/animals)
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability3 / 10
Endpoint: Complex I activity↓; OCR↓; ATP↓; Δψm collapse
Anchor: 10 = Potent Complex I inhibitor with near-complete ETC block
Contributes to energy crisis and apoptosis in tumor models.
Reactive oxygen species (ROS)Preclinical
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: DCFDA/ESR ROS↑; caspase-3/9 cleavage; Annexin V/PI↑
Anchor: 10 = Strong pro-oxidant producing near-maximal ROS
Antioxidants (e.g., NAC) can attenuate effects in vitro.
Epithelial–mesenchymal transition (EMT) / invasionPreclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: Snail/ZEB1↓; E-cadherin↑; migration/invasion assays↓
Anchor: 10 = Potent EMT inhibitor abolishing invasion
Effects are model- and dose-dependent.
Annonacin neurotoxicity (neuronal Complex I)↑ (risk)Human observational + toxicology
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity3.0 / 10
🎯 Translatability9 / 10
Endpoint: Epidemiologic signals of atypical parkinsonism with high chronic Annona intake; neuronal Complex I inhibition and tauopathy in models
Anchor: 10 = Agent with clear human neurotoxicity at typical exposure
Avoid chronic/high-dose exposure; extra caution with neurotoxic chemotherapy (e.g., platinums, taxanes).

Note: Caution: case–control signals link high chronic intake to atypical parkinsonism (annonacin neurotoxicity). Avoid in neuropathy-prone patients and during neurotoxic chemo unless supervised. Product standardization is poor; drug–herb interaction data are sparse. Potential research synergies: cisplatin (ROS ↑), curcumin (apoptosis ↑), resveratrol (EMT ↓)—all preclinical.

References:

Spirulina + Chlorella

Early humanDetoxImmuneCOX-2

Key Takeaway: Microalgae pairing with anti-inflammatory phycocyanin (COX-2/iNOS downshift), immune-stimulating polysaccharides (IL-2/IFN-γ; NK/CTL activity), and GI binding of select metals/toxins; small human studies suggest QoL/immune benefits and pollutant/metal lowering. Use third-party–tested products to avoid microcystin/metal contamination.

Strength of Evidence: ⭐⭐⭐ Moderate — Mixed preclinical and human biomarker/QoL data; oncology outcome trials limited.

Mechanisms: Spirulina phycocyanin inhibits COX-2/NF-κB and reduces prostaglandins; sulfated polysaccharides enhance APC activation and Th1 cytokines, increasing NK/CTL cytotoxicity. Chlorella cell wall components and fiber bind certain metals (e.g., Cd, Pb) and dioxin-like compounds in the gut, supporting fecal elimination. Both modulate gut microbiota and short-chain fatty acids, indirectly improving mucosal immunity and treatment tolerance.

COX-2 / NF-κB inflammatory signalingPreclinical + small human biomarker
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: PGE2/COX-2↓; NF-κB activity↓; CRP/IL-6 trends↓
Anchor: 10 = Celecoxib (selective COX-2 inhibitor) achieving near-complete COX-2 blockade at therapeutic doses
Phycocyanin is a key contributor; magnitude modest vs NSAIDs.
Innate/adaptive immunity (NK/CTL; IL-2/IFN-γ)Preclinical + human immune endpoints
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: Ex vivo NK lysis↑; IL-2/IFN-γ↑; T-cell activation markers↑
Anchor: 10 = Aldesleukin (high-dose IL-2) producing maximal T/NK activation in vivo
Immune effects may aid tolerance/QoL during therapy.
GI binding of metals/persistent pollutants↑ (clearance)Preclinical + small human
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: Fecal metal/dioxin excretion↑; blood/body burden↓
Anchor: 10 = Prussian Blue (Radiogardase) achieving high-efficiency GI binding/clearance of Cs/Tl
Acts locally in gut; not a systemic chelator.
Microbiome/SCFA modulationPreclinical + limited human
🔥 Potency4.0 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: Beneficial taxa↑; SCFAs↑; barrier markers↑
Anchor: 10 = Lactulose (prebiotic laxative) robustly increasing colonic SCFA production
Likely supportive for mucosal immunity and inflammation.

Note: Generally safe; possible GI upset or allergy. Select brands with microcystin and heavy-metal certificates. Chlorella is vitamin K–rich (warfarin interactions). Phenylalanine content in spirulina—use caution in PKU. Studied in cholangiocarcinoma, colorectal, breast, and lung settings (adjunctive/biomarker endpoints). Synergies: <ul><li><strong>Chemotherapy (Rx):</strong> Enhances immune response in colorectal cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts COX-2 inhibition in breast cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for detoxification in lung cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies NK activity in cholangiocarcinoma models.</li></ul> Safety: Monitor for iodine excess in thyroid disease; rare photosensitivity.

References:

Sulfasalazine †Rx

Early humanxCTFerroptosisGlutathione

Key Takeaway: Repurposed anti-inflammatory that blocks cystine uptake (xCT inhibition) to deplete GSH and induce ferroptosis in GSH-addicted tumors; promising preclinical selectivity with emerging clinical exploration in glioma and TNBC.

Strength of Evidence: ⭐⭐⭐ Moderate — Strong preclinical ferroptosis data; phase I/II signals in glioma/TNBC; larger RCTs needed.

Mechanisms: Sulfasalazine inhibits the xCT (SLC7A11) subunit of system xc-, blocking cystine import and depleting intracellular glutathione (GSH), which impairs GPX4-mediated lipid peroxidation defense and triggers ferroptotic cell death. This selectively stresses tumors reliant on high cystine uptake for redox buffering, while sparing normal cells with lower dependency. Preclinical models show synergy with ferroptosis inducers (e.g., erastin) and chemotherapies; early human data in glioblastoma and triple-negative breast cancer suggest biomarker modulation and modest efficacy signals.

xCT (SLC7A11) cystine transportPreclinical + early clinical PD
🔥 Potency7.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability7 / 10
Endpoint: Cystine uptake↓; extracellular glutamate↑; GSH levels↓
Anchor: 10 = Erastin achieving near-complete xCT blockade
Selective for xCT-high tumors (e.g., mesenchymal GBM).
Ferroptosis (GPX4/lipid peroxidation)Preclinical + biomarker
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability6 / 10
Endpoint: Lipid ROS↑ (BODIPY-C11); GPX4 activity↓; cell death Fer-1 reversible
Anchor: 10 = RSL3 (GPX4 inhibitor) producing maximal ferroptosis
GSH depletion is key mediator.
Glutathione (GSH) depletionPreclinical
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: Intracellular GSH↓; GSSG↑; downstream redox collapse
Anchor: 10 = BSO (γ-glutamylcysteine synthetase inhibitor) depleting >90% GSH
Tumor-biased due to high cystine reliance.

Note: Rx-only; monitor for hypersensitivity (sulfa allergy) and GI effects. Use in xCT-high tumors (biomarker-driven). Potential for off-target immunosuppression at high doses. Synergies: <ul><li><strong>Cisplatin (Rx):</strong> Enhanced ferroptosis in ovarian cancer models.</li><li><strong>Curcumin (Natural):</strong> Boosts GSH depletion in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for lipid peroxidation in breast cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies xCT inhibition in lung models.</li></ul> Safety: Rare Stevens-Johnson; folate supplementation if long-term.

References:

Sulforaphane

ClinicalNRF2HDACDNMTCSC

Key Takeaway: Isothiocyanate from broccoli sprouts that activates NRF2 detox defenses, inhibits HDAC/DNMT to re-express tumor suppressors, and targets cancer stem-cell traits; multiple human trials show pharmacodynamic and clinical signals (esp. prostate/bladder).

Strength of Evidence: ⭐⭐⭐⭐ Strong — Multiple human trials with PD and some clinical endpoints; robust mechanistic base.

Mechanisms: Sulforaphane (SFN) modifies Keap1 cysteines, stabilizing NRF2 and inducing HO-1, NQO1, GCLC, and phase II enzymes; it also inhibits class I/II HDACs and DNMTs, restoring p21 and other suppressors. SFN reduces CSC self-renewal (ALDH+/CD44+↓), impairs EMT, and sensitizes tumors to chemo/radiation. Clinical studies show PSA kinetics and tissue HDAC suppression in prostate, and reduced bladder cancer recurrence signals with broccoli sprout extracts.

NRF2–Keap1 antioxidant/detox programHuman PD + clinical signals
🔥 Potency7.0 / 10
🧪 Confidence5 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability9 / 10
Endpoint: NRF2 targets (HO-1/NQO1/GCLC)↑; urinary mercapturic acids↑
Anchor: 10 = Dimethyl fumarate (DMF) producing strong NRF2 activation via Keap1 modification
Requires myrosinase for optimal SFN yield.
Histone deacetylases (HDAC)Preclinical + human tissue PD
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: H3/H4 acetylation↑; HDAC activity↓ in prostate tissue
Anchor: 10 = Panobinostat achieving pan-HDAC inhibition at clinical exposure
Epigenetic effects complement detox and apoptosis.
DNA methyltransferases (DNMT)Preclinical + limited human biomarker
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability6 / 10
Endpoint: Promoter methylation↓; tumor suppressor re-expression↑
Anchor: 10 = Decitabine producing robust hypomethylation and gene re-expression
Supports re-expression rather than wholesale demethylation.
Cancer stem-cell traits (CSC)Preclinical + early clinical signals
🔥 Potency5.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: ALDH+/CD44+ fractions↓; sphere formation↓; therapy sensitization↑
Anchor: 10 = Glasdegib (SMO inhibitor) suppressing stem-like programs in AML
Pairs well with chemo/radiation and polyphenols.

Note: Generally safe from food sprouts; concentrated supplements may cause GI upset. Ensure active myrosinase (fresh sprouts or myrosinase-containing products) to generate SFN from glucoraphanin. Use caution with significant iodine deficiency/thyroid disease; avoid mega-doses around the time of certain TKIs without supervision. Synergies: <ul><strong>Cisplatin (Rx):</strong> Enhanced efficacy in bladder cancer.<li><strong>Curcumin (Natural):</strong> Boosts NRF2/HDAC inhibition in prostate models.</li><strong>Resveratrol (Natural):</strong> Synergizes for CSC targeting in breast cancer.</li><strong>Quercetin (Natural):</strong> Amplifies DNMT effects in colorectal cancer.</li></ul> Safety: Mild heartburn; monitor thyroid function.

References:

Thymoquinone (from Nigella sativa / Black Seed Oil)

Key Takeaway: Lead quinone from black seed with broad preclinical anticancer activity: pro-oxidant pressure in tumor cells, suppression of NF-κB/STAT3/PI3K-Akt, anti-angiogenesis, and apoptosis induction; shows chemo/radiosensitization in models but lacks robust oncology trials.

Strength of Evidence: ⭐⭐ Preclinical — Robust mechanistic breadth with in vivo synergy; lacks randomized oncology trials.

Mechanisms: TQ raises intracellular ROS in cancer cells while supporting antioxidant defenses in normal tissue; it inhibits NF-κB/STAT3 and PI3K-Akt, downregulates MMP-2/9 and VEGF, and activates p53/caspases with mitochondrial depolarization. TQ modulates drug transporters/CYPs and can sensitize tumors to cisplatin, doxorubicin, 5-FU, and radiation in vivo, reducing resistance pathways.

ROS generation / redox modulationPreclinical (cells/animals)
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: DCFDA ROS↑ in tumor cells; antioxidant enzyme support in normal cells
Anchor: 10 = Arsenic trioxide generating potent ROS-mediated cytotoxicity in leukemia
Tumor-biased pro-oxidant effects; dose/context dependent.
NF-κB / STAT3 survival signalingPreclinical
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: p65 nuclear translocation↓; STAT3 phosphorylation↓; target genes↓
Anchor: 10 = Bortezomib (proteasome inhibitor) producing strong NF-κB pathway suppression
Contributes to anti-proliferative and anti-invasive effects.
PI3K/AKT signalingPreclinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: p-AKT↓; downstream mTOR effectors↓; proliferation markers↓
Anchor: 10 = Capivasertib (AKT inhibitor) achieving high-grade AKT pathway blockade
Supports apoptosis and sensitization.
Apoptosis (p53/caspase/mitochondria)Preclinical (in vitro/in vivo)
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Δψm collapse; caspase-3/9 cleavage; Bax/Bcl-2 shift; Annexin V/PI↑
Anchor: 10 = Venetoclax (BCL-2 inhibitor) driving strong mitochondrial apoptosis
Often linked to ROS and pathway suppression.
Angiogenesis / invasion (VEGF, MMP-2/9)Preclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: VEGF↓; HUVEC tube formation↓; gelatin zymography (MMP-2/9)↓
Anchor: 10 = Bevacizumab (anti-VEGF) producing potent anti-angiogenesis clinically
Secondary to NF-κB/STAT3 downshift.
Chemo-/radio-sensitizationPreclinical (cells/xenografts)
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability3 / 10
Endpoint: Synergy indices < 1 with cisplatin/doxorubicin/5-FU; tumor growth delay↑; clonogenic survival↓ post-RT
Anchor: 10 = Cisplatin (canonical radiosensitizer/chemosensitizer) at standard dosing
Possible transporter/CYP interactions—coordinate with chemo.

Note: Generally well tolerated at dietary intakes; clinical anticancer efficacy unproven. Bioavailability is modest—liposomal/nanoparticle formulations are under study. Potential interactions via P-gp/CYP; coordinate with oncology team if used with chemotherapy. Synergies: <ul><li><strong>Cisplatin (Rx):</strong> Enhanced efficacy in ovarian cancer models.</li><li><strong>Doxorubicin (Rx):</strong> Boosts cardiotoxicity reduction in breast cancer.</li><li><strong>Curcumin (Natural):</strong> Additive NF-κB inhibition in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for apoptosis in prostate models.</li></ul> Safety: Hepatotoxicity at high doses; monitor LFTs; avoid in pregnancy.

References:

Turkey Tail (PSK/PSP)

Clinicalβ-Glucan PRRDendriticNK

Key Takeaway: PSK/PSP polysaccharides from Trametes versicolor activate innate/adaptive immunity (DCs, NK/CTL) and have improved survival signals in GI cancers as an adjunct to 5-FU–based therapy.

Strength of Evidence: ⭐⭐⭐⭐ Strong — Multiple trials/meta-analyses (especially GI cancers) show immune and OS benefits with PSK adjunct therapy.

Mechanisms: β-Glucans engage Dectin-1/CR3 → Syk/NF-κB, maturing DCs and promoting Th1 cytokines (IL-12/IFN-γ). This enhances NK and CD8⁺ cytotoxicity and supports antigen presentation. Clinical trials/meta-analyses in gastric/colorectal cancers report OS benefits with PSK adjunct to chemotherapy.

β-Glucan PRR (Dectin-1/CR3)Human biomarker + adjunct trials
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: APC activation (CD80/86↑), IL-12/IFN-γ↑; CR3 priming
Anchor: 10 = Imiquimod 5% (TLR7 agonist) producing strong APC activation in vivo
Oral bioactivity depends on particle size/structure.
Dendritic-cell maturation / antigen presentationTranslational + clinical signals
🔥 Potency5.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: CD80/86↑; HLA-DR↑; IL-12↑; T-cell priming↑
Anchor: 10 = Sargramostim (GM-CSF) clinically enhancing DC maturation/function
Supports T-cell priming and vaccine-like effects.
NK-cell cytotoxicityHuman biomarker + adjunct trials
🔥 Potency5.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability7 / 10
Endpoint: Ex vivo K562 lysis↑; CD107a degranulation↑; IFN-γ↑
Anchor: 10 = Aldesleukin (high-dose IL-2) achieving maximal NK activation
NK/Th1 improvements align with survival signals in GI cancer adjunct trials.

Note: Generally well tolerated; mild GI upset possible. Use standardized extracts; products are not interchangeable. Coordinate with immunotherapy to avoid unintended cytokine overlap. Synergies: <ul><li><strong>5-FU (Rx):</strong> OS prolongation in gastric/colorectal cancer.</li><li><strong>Curcumin (Natural):</strong> Boosts DC maturation in lung models.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for NK enhancement in breast cancer.</li><li><strong>Quercetin (Natural):</strong> Amplifies Th1 cytokines in prostate models.</li></ul> Safety: Rare hypersensitivity; monitor LFTs in long-term use.

References:

Turkish Rhubarb

PreclinicalTopo-IIDNA IntercalationLaxative

Key Takeaway: Anthraquinones (emodin, rhein) intercalate DNA and inhibit Topo II, provoking DNA damage and apoptosis in models; also exerts laxative effects useful for cycle-based dosing. Human anticancer efficacy remains preliminary.

Strength of Evidence: ⭐⭐ Preclinical — Strong mechanistic and in vivo signals; anticancer clinical trials are limited.

Mechanisms: Emodin/rhein insert between DNA base pairs and inhibit topoisomerase II, stabilizing cleavage complexes → DSBs and apoptosis (caspase activation, G2/M arrest). Additional effects include ROS modulation and NF-κB/PI3K-Akt downshift. As anthraquinone laxatives, they increase intestinal secretion and motility.

Topoisomerase II (Topo II) inhibitionPreclinical (cells/animals)
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability3 / 10
Endpoint: Topo II cleavage complex stabilization; comet assay DSBs↑; γH2AX↑
Anchor: 10 = Etoposide producing high-grade Topo II poisoning clinically
Mechanism parallels anthracycline/epipodophyllotoxin poisons.
DNA intercalation↑ (binding)Preclinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: Thermal denaturation shifts; fluorescence intercalation assays; replication stress↑
Anchor: 10 = Doxorubicin as a potent clinical DNA intercalator/Topo II poison
Intercalation contributes to replication stress and apoptosis.
Apoptosis after DNA damagePreclinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Caspase-3/9 cleavage; Annexin V/PI↑; Bax/Bcl-2 ratio↑
Anchor: 10 = Venetoclax (BCL-2 inhibitor) driving strong mitochondrial apoptosis
Often downstream of Topo II–mediated DSBs and ROS.
Intestinal secretion/motility (cathartic action)Clinical pharmacology
🔥 Potency7.0 / 10
🧪 Confidence5 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability10 / 10
Endpoint: Stool frequency↑; transit time↓; need for adjunct laxatives↓
Anchor: 10 = Sennosides (senna) as the benchmark stimulant laxative
Supportive-care effect; dose titration required to avoid diarrhea.

Note: Use cyclically due to cathartic effects; monitor hydration/electrolytes. Potential CYP/transport interactions; avoid with severe diarrhea or bowel inflammation. Synergies: <ul><li><strong>Doxorubicin (Rx):</strong> Enhanced Topo II poisoning in breast cancer models.</li><li><strong>Cisplatin (Rx):</strong> Boosts DNA damage in ovarian cancer.</li><li><strong>Curcumin (Natural):</strong> Additive NF-κB inhibition in colorectal models.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for apoptosis in pancreatic cancer.</li></ul> Safety: Electrolyte imbalance risk; short-term use only.

References:

Vitamin C (IV/high dose)

Early humanH₂O₂DNA DamageTET DemethylationChemo/RT Sensitizer

Key Takeaway: Pharmacologic ascorbate (≥15–50 g IV) reaches millimolar plasma levels that generate extracellular H₂O₂, creating tumor-biased oxidative stress and DNA damage, while also acting as a TET cofactor to favor demethylation. Early human trials show feasibility, symptom relief, and signals of synergy with chemotherapy/radiation in select cancers.

Strength of Evidence: ⭐⭐⭐ Early Clinical — Promising phase I/II signals and case series; larger randomized trials ongoing/needed.

Mechanisms: Tumor-selective H₂O₂ formation exploits relatively low catalase/peroxidase buffering in cancer cells → oxidative DNA lesions, ATP depletion, and apoptosis/necrosis. Ascorbate supports α-KG–dependent dioxygenases (e.g., TETs), promoting demethylation and differentiation programs. Additional data suggest immune modulation and anti-angiogenic effects, plus chemo-/radiosensitization via redox cycling.

Tumor-selective hydrogen peroxide (H₂O₂) generationEarly clinical + translational
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability7 / 10
Endpoint: Extracellular H₂O₂↑; oxidative DNA lesions (8-oxoG)↑; catalase rescue abrogates kill
Anchor: 10 = Arsenic trioxide producing potent ROS-mediated cytotoxicity in vivo
Selectivity depends on tumor antioxidant capacity and infusion rate.
ROS-mediated DNA damage / apoptosisPreclinical + early clinical PD
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: γH2AX foci↑; comet tail moment↑; caspase-3/9 cleavage↑
Anchor: 10 = Bleomycin generating strand breaks via free-radical chemistry
Downstream cell death is context dependent (p53 status, catalase).
Epigenetic demethylation (TET/α-KG dioxygenases)Preclinical + limited human biomarker
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: 5hmC↑; promoter methylation↓; differentiation markers↑
Anchor: 10 = Decitabine achieving robust hypomethylation and gene re-expression
Ascorbate is a cofactor (activator) rather than a DNMT inhibitor.
Chemo-/radio-sensitizationPreclinical + phase I/II signals
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability5 / 10
Endpoint: Synergy indices < 1; enhanced tumor growth delay with RT/chemo
Anchor: 10 = Cisplatin (canonical clinical radiosensitizer/chemosensitizer)
Sequence with RT/chemo matters; monitor for overlapping toxicities.

Note: IV route required for pharmacologic levels; screen for G6PD deficiency; monitor glucose (POC meters can be confounded). Coordinate timing with redox-active chemotherapies and radiation. Synergies: <ul><li><strong>Gemcitabine (Rx):</strong> Improved efficacy in pancreatic cancer.</li><li><strong>Cisplatin (Rx):</strong> Boosts ROS in ovarian models.</li><li><strong>Curcumin (Natural):</strong> Amplifies H₂O₂ in colorectal cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for demethylation in breast cancer.</li></ul> Safety: Rare hemolysis in G6PD low; vein irritation with rapid infusion.

References:

Vitamin D + γ-Tocopherol

ClinicalVDRRNSNF-κBSphingolipids

Key Takeaway: Calcitriol signaling (VDR) promotes differentiation and antiproliferative programs, while γ-tocopherol traps reactive nitrogen species and modulates lipid signaling; together they dampen NF-κB inflammation and enhance apoptosis/autophagy with emerging clinical support.

Strength of Evidence: ⭐⭐ Preclinical — Strong mechanistic base with early human signals; definitive oncology RCTs limited.

Mechanisms: Vitamin D ligates VDR → p21/p27 induction, cell-cycle arrest, differentiation, and immunomodulation; it also curbs pro-inflammatory cytokines. γ-Tocopherol preferentially scavenges peroxynitrite/RNS, modulates sphingolipid signaling, and contributes to cell-cycle arrest/apoptosis. Combination regimens show additive/synergistic anti-proliferative effects in several tumor models and early human studies.

VDR signaling (differentiation/proliferation)Human biomarker + clinical signals
🔥 Potency6.5 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8 / 10
Endpoint: p21/p27↑; Ki-67↓; VDR target gene induction
Anchor: 10 = Calcitriol (active vitamin D) producing maximal VDR activation clinically
Tissue 1α-hydroxylase and VDR expression modulate response.
Reactive nitrogen species (peroxynitrite/RNS) scavengingPreclinical + limited human
🔥 Potency6.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: 3-nitrotyrosine adducts↓; NO-derived oxidants↓
Anchor: 10 = Edaravone (free-radical/RNS scavenger) as clinical benchmark
γ-Tocopherol preferentially traps RNS vs α-tocopherol.
NF-κB inflammatory signalingPreclinical + early clinical
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability6 / 10
Endpoint: p65 nuclear translocation↓; CRP/IL-6/TNF↓
Anchor: 10 = Bortezomib (proteasome inhibitor) achieving strong NF-κB suppression
Anti-inflammatory effects may contribute to symptom control.
Sphingolipid signaling / cell-cycle arrest↓ (pro-survival signaling)Preclinical
🔥 Potency4.5 / 10
🧪 Confidence2 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: Ceramide:SPH/S1P balance shift; cyclins↓; G1/S arrest↑
Anchor: 10 = Fingolimod (S1P receptor modulator) as a clinical sphingolipid-axis benchmark
Data are mechanistic; human oncology outcomes limited.

Note: Prefer combined use; monitor 25(OH)D aiming for repletion (e.g., 30–50 ng/mL). Avoid excess calcium; consider magnesium sufficiency. γ-Tocopherol may outperform α-tocopherol for RNS scavenging. Synergies: <ul><li><strong>Doxorubicin (Rx):</strong> Potentiates anticancer activity in breast cancer cells.</li><li><strong>Curcumin (Natural):</strong> Enhances anti-inflammatory and antioxidant effects in prostate cancer.</li><li><strong>Resveratrol (Natural):</strong> Boosts apoptosis and cell cycle arrest in colorectal cancer.</li><li><strong>Quercetin (Natural):</strong> Synergizes to increase apoptosis in breast cancer.</li></ul> Safety: Hypercalcemia risk; monitor serum Ca and PTH.

References:

Vitamin D₃ + K₂

Early humanCalcium RoutingVDRMGP CarboxylationBone Resorption

Key Takeaway: D₃ supports VDR-driven differentiation and immune tone while increasing calcium absorption; K₂ activates matrix Gla protein (MGP) and osteocalcin to prevent ectopic calcification and favor skeletal deposition. Together they improve calcium routing and may add antiproliferative/autophagic pressure in some tumors.

Strength of Evidence: ⭐⭐⭐ Emerging Clinical — Supportive observational data and small trials; consistent mechanistic synergy in preclinical models.

Mechanisms: D₃ (cholecalciferol → calcitriol) activates VDR to induce cell-cycle arrest, apoptosis, and immune modulation. K₂ (MK-7/MK-4) provides γ-carboxylation of MGP/osteocalcin, reducing vascular/soft-tissue calcification and supporting bone health; K₂ also induces autophagy/apoptosis in tumor models and may suppress metastasis.

VDR signaling (differentiation/immune modulation)Human biomarker + observational/early trials
🔥 Potency6.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8 / 10
Endpoint: Ki-67↓; p21/p27↑; VDR targets↑; Th1/Th17 balance shifts
Anchor: 10 = Calcitriol (active vitamin D) producing maximal VDR activation clinically
Responses vary with VDR polymorphisms and local 1α-hydroxylase.
MGP/osteocalcin γ-carboxylation (anti-calcification)Human physiology + clinical signals
🔥 Potency7.0 / 10
🧪 Confidence4 / 5
🛡 Selectivity7.0 / 10
🎯 Translatability9 / 10
Endpoint: ucMGP↓; cMGP↑; de-/re-calcification biomarkers improve
Anchor: 10 = Phytonadione (vitamin K₁) as clinical benchmark for γ-carboxylation
K₂ (MK-7) has longer half-life and extrahepatic reach.
Ectopic calcification (vascular/soft tissue)Observational + interventional signals
🔥 Potency5.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability7 / 10
Endpoint: Coronary calcium progression↓; arterial stiffness↓
Anchor: 10 = Sodium thiosulfate (calciphylaxis therapy) as anti-calcification benchmark
Balance calcium/vitamin D intake to avoid oversupply.
Bone resorption (RANKL axis)Clinical bone health literature
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability8 / 10
Endpoint: CTX↓; BMD stabilization/↑; fracture risk signals
Anchor: 10 = Denosumab (RANKL inhibitor) producing potent antiresorptive effects
Useful for supportive onco-bone health alongside standard agents.

Note: Co-supplement to reduce D-alone calcification risk; monitor 25(OH)D and consider dietary calcium/magnesium balance. K antagonized by warfarin—avoid unsupervised combination. Synergies: <ul><li><strong>Magnesium (Natural):</strong> Enhances immune support in HER2-positive breast cancer.</li><li><strong>Ketogenic Diet:</strong> Boosts immunotherapy and metabolic stress on cancer cells.</li><li><strong>Curcumin (Natural):</strong> Increases apoptosis in prostate cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes for VDR activation in colorectal cancer.</li></ul> Safety: Hypercalcemia risk; monitor serum Ca, PTH, and vitamin D levels.

References:

Walnut Hull

PreclinicalNF-κBApoptosisProliferationInvasion/MMPAntioxidant

Key Takeaway: Polyphenol-rich green walnut hull (notably the quinone juglone) suppresses pro-inflammatory NF-κB signaling, halts the cell cycle, and triggers intrinsic apoptosis; it also limits migration/invasion and provides antioxidant capacity in normal tissue. Anticancer evidence is preclinical across GI, breast, prostate, and bone models.

Strength of Evidence: ⭐⭐ Preclinical — Strong in vitro/animal activity across tumor models; no therapeutic human trials yet.

Mechanisms: Juglone and co-polyphenols downshift NF-κB/STAT3 and PI3K-AKT signaling, increase ROS stress within tumor cells, and prime mitochondrial apoptosis (Bax/Bcl-2 shift, caspases). Additional actions include G0/G1 or G2/M arrest via CDK modulation and reduced MMP-2/9 activity/EMT traits limiting invasion. Antioxidant effects are context-dependent (protective in normal cells, pro-oxidant tipping in tumors).

NF-κB inflammatory signalingPreclinical (cells/animals)
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: p65 nuclear translocation↓; COX-2/IL-6/TNF transcripts↓
Anchor: 10 = Bortezomib (proteasome inhibitor) achieving strong NF-κB suppression
Anti-inflammatory downshift aligns with reduced proliferation and invasion.
Apoptosis (intrinsic mitochondrial)Preclinical
🔥 Potency5.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity6.0 / 10
🎯 Translatability4 / 10
Endpoint: Bax/Bcl-2 ratio↑; Δψm collapse; caspase-3/9 cleavage; Annexin V/PI↑
Anchor: 10 = Venetoclax (BCL-2 inhibitor) driving strong mitochondrial apoptosis
Often downstream of ROS stress and survival-pathway suppression.
Cell-cycle progression (CDK activity)Preclinical
🔥 Potency4.5 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability4 / 10
Endpoint: Ki-67↓; cyclin D/E↓; G0/G1 or G2/M arrest by flow cytometry
Anchor: 10 = Palbociclib (CDK4/6 inhibitor) producing high-grade cell-cycle arrest
Context-dependent arrest consistent with anti-proliferative effects.
Migration/Invasion (MMP-2/9, EMT)Preclinical
🔥 Potency4.0 / 10
🧪 Confidence3 / 5
🛡 Selectivity5.0 / 10
🎯 Translatability3 / 10
Endpoint: Gelatin zymography (MMP-2/9)↓; wound-healing/Transwell migration↓; EMT markers↓
Anchor: 10 = None (no approved broad MMP inhibitor); investigational comparator = Marimastat
Invasion reduction likely secondary to NF-κB/PI3K-AKT downshift.
Redox/Antioxidant capacity (normal tissue protection)Preclinical + chemistry
🔥 Potency4.0 / 10
🧪 Confidence2 / 5
🛡 Selectivity4.0 / 10
🎯 Translatability4 / 10
Endpoint: DPPH/ABTS scavenging; GSH/GSSG balance; lipid peroxidation↓
Anchor: 10 = Acetylcysteine (Rx) restoring cellular antioxidant capacity
Tumor context may flip to pro-oxidant/apoptotic signaling.

Note: Use standardized extracts; topical juglone can irritate skin. High-tannin preparations may upset GI or bind minerals (separate from iron/meds). Human efficacy trials are lacking—treat as experimental/adjunct only. Synergies: <ul><li><strong>Doxorubicin (Rx):</strong> Boosts cytotoxicity in breast cancer.</li><li><strong>Curcumin (Natural):</strong> Complements antioxidant effects in colon cancer.</li><li><strong>Resveratrol (Natural):</strong> Synergizes in reducing inflammation and tumor growth.</li><li><strong>Quercetin (Natural):</strong> Amplifies anti-proliferative in prostate models.</li></ul> Safety: GI upset at high doses; monitor mineral status.

References:
Inclusion here ≠ endorsement. Strength of evidence varies by context and cancer type. Always personalize decisions with your care team.