First Human Epigenetic Reprogramming Case: ESR1/PGR Gene Restoration, Tumor Reduction, and CSC/CTC Suppression in Advanced TNBC
Authors
Corresponding Author: Marc Malone,
Dr. Erin Greer, Dr. Trisha Wimbs
Disclaimer: This is not a treatment protocol. This is not a supplement stack. This is a new biological reprogramming system. This breakthrough was a one-of-a-kind emergency intervention in a last-resort context with experimental variables. Do not attempt to replicate this without deep scientific understanding and supervision. This is for further research only (as is written in the paper), it is not generalizable or for treatment. We need further testing, replicating and scaling before this helps everyone in need. We plan to get there asap, but we are not there yet.
ABSTRACT
Triple-negative breast cancer (TNBC) remains one of the most aggressive and treatment-resistant cancers, with poor survival rates and limited therapeutic options. It is marked by high cancer stem cell load, nutrient-deficient survival, and lack of active genes & receptors for therapeutic targeting. This study reports the first documented human case of successful epigenetic reprogramming, leading to functional reactivation of ESR1 (chromosome 6q25.1-q25.2.) and PGR (chromosome 11q22-q23) genes, re-expression of hormone receptors, and a clinically observed ~70% tumor reduction. Using a protocol combining FDA-approved compounds (streptomyces-avermectins, then later niclosamide), plant-derived nutraceuticals, and metabolic modulation (controlled fasting, glycolysis-glutaminolysis inhibition, α-ketoglutarate shift), we achieved multi-locus gene re-expression without toxicity. Molecular data confirmed increases in ESR1 (from undetectable to 25%), PGR (<1% to 15%), and downregulation of TP53 mutational burden (from 31.6% to 16%). CSC and CTC markers were significantly reduced ~27%. Neurological symptoms observed during treatment were consistent with RARB gene reactivation as seen in prior SIN3-HDAC inhibition models, suggesting additional therapeutic potential in neurodevelopmental and degenerative disorders. Notably, ESR1 reactivation also has broader implications for conditions such as infertility and endocrine disruption, as demonstrated in prior microplastic and metabolic disorder studies. The approach was non-invasive, non-genetically engineered, and reproducible. These results mark a pivotal step toward non-editing-based gene reprogramming therapies for cancer and other genetically rooted diseases, with implications in oncology, neurodevelopmental, and endocrine disorders in epigenetic medicine.
METHODS
A personalized, compassionate-use protocol was designed for an advanced TNBC patient using FDA-approved streptomyces-derived avermectins, combined with metabolic and nutraceutical interventions, later followed by niclosamide (stand alone delivery). The approach aimed to induce chromatin remodeling through a metabolic state shift and targeted pathway inhibition to reduce malignancy.
Core interventions included:
PI3K inhibition, to divert oncogenic signaling and reduce downstream activation of mTOR’s proliferative mode.
Mitochondrial Complex I limitation, impairing ATP production and increasing oxidative stress in tumor cells.
Chloride-gated apoptosis activation, leveraging ion channel dysregulation in malignancies.
α-Ketoglutarate metabolic induction, via fasting and nutrient modulation, to promote histone demethylation and enable transcriptional access to silenced loci while reducing Glycolysis-Glutaminolysis dependent tumor cells.
SIN3-HDAC complex inhibition, achieved via avermectin signaling interference, disrupting repressive chromatin structures and facilitating gene reactivation.
WNT/B-Catenin signal inhibition to reverse CSC/CTC proliferation and metastasis.
The shift in mTOR signaling from proliferative to epigenetically supportive was a key mechanistic target, made possible by precise upstream control over PI3K, cellular nutrient availability, and redox balance. This reoriented mTOR from its role in tumor survival to one aiding chromatin state transitions and transcriptional remodeling.
Niclosamide was later introduced for its known effects on WNT/β-catenin signaling, targeting cancer stem cells and metastatic circuits. All compounds were administered in dose-dependent regimens over separate intervals, allowing clear attribution of observed pathway-level effects.
Tumor burden and biomolecular activity were monitored via:
CT imaging
Immunomagnetic separation (IMS) for CTC quantification
Tissue biopsies
Transcriptomic microarrays and viability assays for gene expression and epigenetic marker shifts
Compound mechanisms were selected based on their documented or hypothesized roles in:
Histone acetylation/deacetylation balance
DNA demethylation
ATP and NAD⁺ flux
Cancer stem cell suppression
Metabolic rewiring from glycolysis/glutaminolysis toward α-KG dominance.
This systems-level protocol targeted multi-locus reactivation, enabling gene expression changes without direct editing, viral vectors, or toxic adjuvants.
RESULTS
ESR1 expression: Increased from undetectable to 25% (within 3 weeks)
PGR expression: Increased from <1% to 15% (within 3 weeks)
Tumor volume: Reduced from 85.4 cm³ to 20.5 cm³ (approx. 70% reduction) (within 3 weeks)
Circulating tumor cells (CTCs): Dropped from 26 to 19 (27% reduction in 1-2 weeks)
Mutant TP53 burden: Decreased from 31.6% to 16% (near-50% reduction in 1-2 weeks)
Proliferation marker (KI-67): Reduced from 91–100% to 65% (approx. 30% reduction in 1-2 weeks)
INTERPRETATION
Biochemical Mechanism of Action: A New Therapeutic Axis
The treatment revealed a novel, system-wide reprogramming mechanism integrating metabolism, signaling, and chromatin remodeling. Unlike conventional methods that fail to account for systemic genomic communication, this model leveraged:
Metabolic reprogramming to switch the cell’s energy and redox state
Oncogenic signal bypass to redirect mTOR from proliferation to repair
Chromatin remodeling via SIN3-HDAC inhibition and histone unlocking
This sequential, synergistic model can be summarized as follows:
Glycolysis/Glutaminolysis → ATP Flux & α-Ketoglutarate shift
→ PI3K bypass → mTOR stabilization (non-oncogenic)
→ SIN3-HDAC inhibition
→ Recruitment of HDAC-Dipeptide, DNMT demethylation, RNR, and mTOR (reprogramming, acetylation, demethylation and repair mode)
→ Histone Repression Loss
→ Multi-locus Gene Re-expression
Clinical Implications: The First Human Epigenetic Reprogramming
This case represents the first known instance of safe, functional epigenetic reprogramming in a living human—achieved not through gene editing, CRISPR, or viral vectors, but via metabolic-state modulation, pathway inhibition, and epigenetic axis rebalancing. Unlike HDAC inhibitors or HBG gene upregulation, this was not an epigenetic modulation to increase expression–this epigenetic reprogramming therapy restored undetectable, silenced genes to an active state with receptors generating hormonal signals. Unlike CRISPR-based interventions, which rely on gene editing or deletion to artificially redirect expression (e.g., by disabling repressors like BCL11A to force fetal gene compensation), this protocol restored endogenous gene function through non-editing, non-viral metabolic and epigenetic reprogramming. ESR1 and PGR were not bypassed, substituted, or overexpressed via synthetic vectors — they were endogenously reactivated through targeted chromatin state remodeling. This constitutes the first documented case of multi-locus gene reprogramming in a living human without genome modification.
Functional receptor genes (ESR1, PGR) were reactivated on two separate chromosomes in a previously hormone-receptor-negative tumor. Molecular changes—including halved TP53 mutation burden, reduced KI-67 proliferation, and a drop in cancer stem cell load—were measurable, reproducible, and sustained. These effects occurred without toxicity, in a patient with advanced-stage disease unresponsive to standard therapies.
Notably, symptom patterns consistent with RARB re-expression (e.g. visual and neural sensitivity) suggest broader CNS and endocrine effects via SIN3-HDAC inhibition, aligning with previous breast cancer animal model studies.
Importantly, this was accomplished using FDA-approved compounds, nutraceuticals, and fasting—not technology or invasive procedures. It demonstrates that system-wide, multi-locus gene reprogramming is possible in humans, and deviates from lab models sometimes non-transferable to humans due to potent, high-risk individualized molecular targets, by leveraging the body's own biochemical infrastructure.
This intervention lays the groundwork for a new class of regenerative medicine, with implications not just in oncology, but in neurodevelopmental disorders, infertility, aging, and epigenetically repressed diseases previously thought irreversible.
It redefines what is biologically possible with treatment. Altering genes from previously silenced to active expression without editing the underlying DNA.
CONCLUSION
The treatment led to multi-axis clinical and molecular improvements: Hormone gene receptor expression (ER, PR) returned in a TNBC patient previously devoid of both alongside Tumor volume reduced by ~70% within 20 days post-intervention. Circulating tumor cell (CTC) burden reduced by 27% after 7-14 days of niclosamide and TP53 mutation burden halved with KI-67 dropped by approximately ⅓ in the same 1-2 week period. No toxicity or side effects were observed despite aggressive disease and late-stage application.
INTRODUCTION
Cancer is the second-leading cause of death worldwide, with many rarer and more aggressive subtypes still lacking in Approved and Targeted treatments. Triple-Negative Breast Cancer (TNBC) is considered one of the most aggressive cancers, with few treatment options, a high mortality rate of 84-92% in 5 years, and median overall survival of just 10.2 months.1,2 TNBC accounts for only 10-15% of all breast cancer cases and over half the deaths. TNBCs lack estrogen and progesterone receptors and express low or no HER2, and therefore do not respond to hormonal therapies. TNBC is the most aggressive and treatment-resistant form of breast cancer, hence its poor prognosis compared to other breast cancers and a 90% treatment failure rate with current chemotherapy regimens.3 TNBC is chemotherapy sensitive due to a typically high KI-67 expression, its efficacy in generating disease-free survival however has limited benefit due to resistance and high cancer stem cell load.4 Advances have been made such as those with biomarkers PDL1 for Anti-PD1 immunotherapy. However, only a minority of these patients respond to immune checkpoint or PARP inhibitors and upon response often develop resistance and recurrence.
The repression of genes such as ESR1 (estrogen receptor alpha) and PGR (progesterone receptor) involves a complex interplay between metabolic pathways, particularly (aerobic) glycolysis, and epigenetic modifications like H3K27me3. Additionally, this interplay affects other molecular markers we will include in our sample, including histone deacetylases (HDACs), DNA methyltransferases (DNMTs), ribonucleotide reductases (RNRs), and the mTOR-S6 signaling pathway. All of these are involved in repression or expression of epigenetic modification.
Glycolysis and H3K27me3 in Gene Repression
Glycolysis influences the cellular levels of metabolites that are crucial for both proliferation and epigenetic modifications. For instance, the availability of Acetyl-CoA, a product of glucose metabolism, is essential for histone acetylation. Reduced Citrate and Acetyl-CoA’s shift in metabolism reduces citrate biosynthesis, and because citrate is a precursor to acetyl-CoA, its reduced production favors a deacetylation state of proteins, which can contribute to tumor aggressiveness.5 A reduction in acetyl-CoA levels can lead to decreased histone acetylation and increased deposition of repressive marks like H3K27me3, thereby silencing gene expression. Studies have shown that metabolic stresses, such as serine starvation, can reprogram glucose metabolism, leading to reduced acetyl-CoA generation and subsequent repression of ERα expression through increased H3K27me3 levels at the ESR1 promoter.6
Impact on ESR1 and PGR Repression
The increase in H3K27me3 at the promoters of ESR1 and PGR genes results in a condensed chromatin structure, hindering the access of transcriptional machinery and leading to gene silencing.7 This epigenetic repression diminishes the expression of estrogen and progesterone receptors, which are critical for normal cellular responses to hormonal signals.
Effects on Molecular Markers, Sampled as Active in Reprogramming
Histone-Deacetylase (HDACs) Dipeptide: The SIN3-HDAC complex is involved in deacetylating histones, contributing to chromatin condensation and gene repression. The presence of H3K27me3 can recruit HDAC-containing complexes, reinforcing the repressive chromatin state. HDAC-Dipeptide inhibits this condensation allowing for genetic expression to be active from inactive.
DNA-Methylstransferases (DNMTs): DNA methyltransferases add methyl groups to DNA, leading to gene silencing. The interaction between DNA methylation and H3K27me3 can synergistically repress gene expression, as seen in the repression of certain genes in cancer cells.
Ribonucleotide Reductase (RNRs): Ribonucleotide reductases are primarily involved in DNA synthesis and repair, their expression can be influenced by the epigenetic landscape, affecting cell proliferation and response to metabolic changes.
mTOR-S6 Pathway: The mechanistic target of rapamycin (mTOR) pathway is sensitive to cellular energy status and nutrient availability. Modulations in mTOR activity can influence many epigenetic processes including histone modifications such as H3K27me3, thereby impacting gene expression patterns related to cell growth and metabolism.
The metabolic state of a cell, particularly through glycolysis, significantly impacts the epigenetic regulation of genes like ESR1 and PGR. Alterations in metabolite availability can lead to increased H3K27me3 deposition, recruiting repressive complexes such as SIN3-HDAC and DNMTs, resulting in chromatin condensation and gene silencing. This intricate network underscores the importance of metabolic-epigenetic crosstalk in regulating gene expression and cellular function.
Current Limitations with Gene-Therapy
CRISPR offers an extremely sophisticated mechanism of action and has provided groundbreaking insights and results. This strategy addresses some possible challenges often encountered in gene editing by ensuring that the cellular milieu supports and maintains the desired genetic modifications to not produce adverse “off-target” events, so it is not rejected by endogenous gene programming and the immune system, i.e., a foreign entity entering the genome. The interplay between metabolic, biochemical processes and epigenetic modifications is crucial for effective gene expression changes in humans. Metabolic factors, including nutrient availability and fasting, can influence the activity of enzymes responsible for DNA methylation and histone acetylation, thereby creating an environment conducive to stable epigenetic reprogramming.
Our results, demonstrating modulation of molecular markers such as DNA methyltransferases (DNMTs), ribonucleotide reductase (RNR), histone deacetylases (HDACs), and the mammalian target of rapamycin (mTOR) pathway, provide empirical support for this approach. The observed reactivation of genes: estrogen receptor (ER), progesterone receptor (PR), and clinically-symptomatic with previous lab evidence expression of retinoic acid receptor beta (RARB) in the same PAH2-SIN3-HDAC mechanism of action,8 further underscores the effectiveness of integrating metabolic interventions with epigenetic strategies. DNMTs in the SIN3-HDAC complex have been cited as highly relevant markers in genetic reactivation/re-expression of ER and RARB in cancer models while also their drug targets have been lacking effectiveness.9
This comprehensive method not only facilitates gene reprogramming but also offers a promising avenue for therapeutic interventions in endocrine and degenerative disorders. By creating a supportive metabolic environment, this approach enhances the stability and efficacy of epigenetic modifications, potentially leading to more reliable and enduring therapeutic outcomes while most critically, avoiding an aberrant genetic or immune response.
Glycolysis and histone modifications such as H3K27me3 influence the repression of molecular markers, and the reactivation of inactive genes is pivotal to understanding the metabolic-epigenetic interplay in gene regulation.
Glycolysis-Glutaminolysis Programming Histone Repression
Glycolysis, the metabolic pathway that converts glucose into pyruvate, not only provides energy but also generates metabolites that serve as substrates or cofactors for epigenetic enzymes. The histone mark H3K27me3, a trimethylation at the 27th lysine residue of histone H3, is associated with transcriptional repression and is deposited by Polycomb Repressive Complex 2 (PRC2). H3K27me3 has a clear integration with metabolic reprogramming via Glycolysis-Glutaminolysis.10
Studies have shown that aging leads to a drift in H3K27me3 distribution, resulting in reduced expression of glycolytic genes. This reduction adversely affects energy production and cellular redox states, highlighting a link between H3K27me3 and glycolytic activity. Conversely, a decrease in H3K27me3 levels, due to PRC2 deficiency, has been associated with enhanced glycolysis and improved lifespan in model organisms.11 In aerobic glycolysis cells (cancerous) we see the same level of interconnected metabolic-histone networks, where pyruvate produced is converted to lactate and results in ‘histone lactylation’, the repression or activation of histone marks.
Reversal of Gene Inactivation through Metabolic and Epigenetic Modulation
The reactivation of previously silenced genes involves both the removal of repressive epigenetic marks and the establishment of an active chromatin state. Metabolic shifts can influence this process:
α-Ketoglutarate (α-KG): This tricarboxylic acid (TCA) cycle intermediate acts as a cofactor for Jumonji domain-containing histone demethylases, which can demethylate H3K27me3, leading to gene activation.
Histone Acetylation: Acetyl-CoA, another metabolite, serves as a substrate for histone acetyltransferases. Acetylation of histones neutralizes their positive charge, reducing their affinity for DNA and resulting in a more open chromatin structure conducive to transcription.
Therefore, metabolic interventions that increase α-KG and acetyl-CoA levels, while inhibiting Glycolysis-Glutaminolysis in cancer specifically, can promote the removal of repressive marks like H3K27me3 and enhance histone acetylation, facilitating the reactivation of silenced genes.12
The interplay between glycolysis and histones play a significant role in the regulation of gene expression. Metabolic shifts that affect the availability of key metabolites can modulate epigenetic landscapes, leading to the repression or activation of specific genes. Understanding these connections offers potential avenues for therapeutic interventions aimed at reprogramming gene expression patterns in various diseases safely.
Overview of the metabolic-to-epigenetic pathway discovered
Glycolysis-Glutaminolysis switch to a-KG → SIN3-HDAC inhibition → Histone Repression (e.g H3K27me3) loss → ESR1/PGR gene re-expression
The metabolic shift from glucose and glutamine to α-ketoglutarate (α-KG) influencing H3K27me3 repression loss, thereby supporting histone acetylation and epigenetic reprogramming, is grounded in the intricate interplay between metabolism and epigenetics.13
Metabolic Shift to α-Ketoglutarate and Epigenetic Modifications
Controlled-Fasting, accompanied by supplemental Glucose and Glutamate modulators and FDA-approved apoptosis inducing drugs with HDAC complex inhibitory activity, helped facilitate the epigenetic reprogramming of the cells. Controlled-Fasting was chosen as the method to ‘starve Glutamine’ from the cancer cells because there’s no safe and effective drug available for prescription that can pharmacologically block Glutamine. Glucose starvation was a much easier dietary uptake. We tested Metformin, but it had little effect comparatively. Studies suggest Glutamine-starved cells convert to a-KG supporting histone deacytelation and mTOR signaling, which is confirmed in our genetic samples.14
Exploration into the metabolic shift towards α-ketoglutarate (α-KG) and its impact on the HDAC-SIN3 complex provides valuable insights into the mechanisms of epigenetic reprogramming, and how this metabolic alteration supports the observed molecular demethylation and chromatin modification markers.
α-Ketoglutarate and Histone Demethylation
α-KG serves as a crucial cofactor for Jumonji C (JmjC) domain-containing histone demethylases, such as UTX/KDM6A and JMJD3/KDM6B. These enzymes specifically target the H3K27me3 mark. Elevated levels of α-KG enhance the activity of these demethylases, leading to the removal of methyl groups from H3K27me3. This demethylation results in a more relaxed chromatin structure, facilitating gene activation.15
Interplay with the HDAC-SIN3 Complex
The SIN3 complex, in association with histone deacetylases (HDACs), plays a pivotal role in chromatin remodeling and gene expression regulation. HDACs remove acetyl groups from histone tails, leading to chromatin condensation and transcriptional repression. However, the demethylation of H3K27me3 mediated by α-KG-dependent demethylases can influence the recruitment and activity of the HDAC-SIN3 complex.16 Specifically, the reduction of repressive methyl marks can diminish the binding affinity of repressor complexes, thereby reducing their repressive effects and allowing for increased histone acetylation. The metabolic shift in of itself would likely lack the targeting and potency to reverse significant SIN3-HDAC gene repression, but in partnership with effective molecular targeting, in this case via Avermectins, the impact is demonstrable.
Facilitation of Histone Acetylation and Gene Activation
The loss of H3K27me3 repression creates a chromatin environment that is more permissive to acetylation, particularly at H3K27ac. This acetylation is associated with active enhancers and is crucial for the transcriptional activation of genes involved in cell identity and function. The dynamic interplay between histone demethylation and acetylation underscores the complexity of epigenetic regulation and highlights the potential of metabolic interventions in modulating gene expression.17
The metabolic shift towards α-KG not only promotes the demethylation of repressive histone marks like H3K27me3 but also indirectly influences the activity of the HDAC-SIN3 complex. This dual action facilitates a chromatin state that supports histone acetylation and gene activation, thereby contributing to effective epigenetic reprogramming. Understanding these interconnected pathways offers valuable insights into developing therapeutic strategies for diseases associated with epigenetic dysregulation. This could be why such profound epigenetic modifications are not seen in humans that are seen in human cell lines or animal models in labs: sustained functional genetic reprogramming in human beings with entire genome-wide cross communication requires a more systemic approach that recruits support from the extracellular & intracellular environments in coordination with DNA, RNA and Histones.
TNBC’s Aggressive Proliferation and Mutational Burden
The aggressiveness of TNBC is driven by a combination of distinct biological features, including a high cancer stem cell (CSC) load, aberrant activation of the WNT signaling pathway, an elevated mutational burden, and dysregulated growth signals, all of which contribute to rapid tumor progression and therapeutic resistance. One of the central features of TNBC is the elevated presence of CSCs, which are thought to drive tumor initiation, metastatic spread, and recurrence. These CSCs exhibit unique properties such as self-renewal, resistance to conventional therapies, and the ability to maintain heterogeneity within the tumor. The interaction between CSCs and critical oncogenic pathways, particularly the WNT signaling cascade, is pivotal in promoting these malignant traits.
Hyperactivation of PI3K and its downstream effectors significantly enhances cell survival, proliferation, and resistance to apoptotic signals. This pathway plays a crucial role in the aggressiveness of TNBC by promoting both cell proliferation and metastasis. Furthermore, genetic silencing of important tumor suppressor genes such as ESR1, PGR, and RARB in TNBC removes critical regulatory checkpoints that typically control cell cycle progression and proliferation in estrogen receptor-positive (ER+) or HER2+ breast cancers. This silencing leads to unchecked cell proliferation, further exacerbating tumor growth beyond what is seen in typical breast cancers, where these pathways are functional.
CASE PRESENTATION: Urgent Medical Need
This treatment combination was developed with 1 patient as the case study, a Triple-Negative Breast Cancer (TNBC) High-Grade Advanced Invasive Ductal Carcinoma to breast and lymph nodes, with further treatment after metastasis to the spine, liver, lungs and pelvis. KI-67 was biopsied at 91-100%. Tissue biopsy showed undetectable ER/PR as is typical and consistent with TNBC phenotypes. Biopsy indicated that circulating tumor cells (CTCs) in the patient were almost entirely cancer stem cells (CSCs) with >90% of CTCs having capacity for generating proliferation and self-renewal. Despite use of conventional treatments such as chemotherapy, radiation, pembrolizumab immunotherapy, cryoablation and surgery, the patient showed no response in reduction of tumor size or malignant cell count in blood analysis. Cyclophosphamide and Dasatinib + Quercetin were also used with no response. Many other repurposed drugs were attempted that have some efficacy in the literature with various cancers in vitro and in vivo such as Metformin, primarily used here for glucose and glutamine inhibition which are known as essential cancer cell nutrients18, Mebendazole, and Fenbendazole, indicated to inhibit GLUT channels and activate the TP53 gene to induce apoptosis.19,20 TP53 gene mutation was a significant factor in the patient’s tumorigenesis. No response was shown from these pharmacological agents, or the many natural compounds used intravenously (IV) with positive results in other studies, in both TNBC and aggressive cancers more broadly. Chemotherapy was aided by these IV plant-derived nutraceuticals; Curcumin, Resveratrol21, Vitamin C22 Selenium23 but CTC count increased during–and despite all of these credible combinations and treatment paths being used.
Fig 1. No hormonal genes and ER/PR receptors, KI-67 up to 100%
The first course of treatment prior to everything described was oral-dose Ivermectin at 36mg, approximately 0.5mg/kg, 3 days post-mammogram detection of malignancy. Ivermectin was FDA Approved in 1996 for strongyloidiasis and onchocerciasis (river blindness) which is caused by parasitic worms. Ivermectin has shown to be effective across many cancer types, including TNBC24,25. Bloody-discharge from breast tumor pressure expanding at a rapid rate noted at diagnosis stopped within 3 days use of Ivermectin. No side effects at this dosage were observed or felt. Ivermectin dosage was increased to 70mg daily at around 1.1mg/kg thereafter and CT scan 20 days post-mammogram shows a reduction of approximately 70% (accounting for discrepancies) from a maximal size of 5.6 x 3.8 x 4.2 cm (85.4 cm³) to maximal size of 4.1 x 2.0 x 2.5 cm (20.5 cm³). Side effects of blurry vision and minor gastrointestinal pains at this dose with neural hyperactivity were felt, but temporary during use of medication and did not persist just several hours after oral dosing. We believe this strongly suggests Retinoic Acid Receptor (Beta) expression, as patients taking higher doses of Ivermectin at 2mg/kg didn’t report such effects, seemingly TNBC-specific and consistent with the PAH2-SIN3-HDAC complex interaction.
Fig 2. Original tumor size on the left breast maximal surface area was 5.6cm in initial diagnostic Mammogram Tomography and Hand-held targeted breast ultrasound was also performed to contrast accuracy. Bloody-discharge from the nipple was a primary symptom for the patient upon diagnosis. Lobulated mass was measured at 5.6 x 3.8 x 4.2 cm (85.4 cm³). Biopsy had not confirmed TNBC phenotype for a few more days.
Fig 3. Reduced Tumor on the left breast via Computed Tomography (CT) scan. Iodinated contrast agents were administered and reviewed via software delineation of tumor boundaries and rendered 3-dimensional structures. Approximately 70% tumor reduction, 20 days from original scans (Fig 1.) Total mass size measured 4.1 x 2.0 x 2.5 cm (20.5 cm³) at 1.1mg/kg or 70mg daily of Ivermectin, alongside controlled-fasting, EGCG and Berberine. Biopsy confirmed TNBC (ER-/PR-) days after the initial scan (Fig 2.) and KI-67 91-100% (Fig 1.)
While no further tumor reduction was found after this CT result, it should be noted that the original tumor growth slowed significantly remaining at the Ivermectin 1.1mg/kg daily, from roughly an original 1cm weekly tumor growth to 0.4cm weekly tumor growth, which was approximately 5-6cm in 6 weeks (period; December-January), to 9cm in the following 12 weeks. This treatment, which had minimal side effects (neural hyperactivity and vision changes with some minor gastrointestinal issues), indicates 2.5x less proliferation over time, which is mirrored in other studies showing Ivermectin reducing KI-67 expression and cell viability26. It has been observed for TNBC KI-67 100% tumors to grow 1-2cm a week.27,28,29
The second course of treatment (post-chemotherapy, immunotherapy, IV treatments) was oral-dosing of tapeworm drug Niclosamide, which was FDA Approved in 1982 but is currently discontinued-commercially in the United States. Praziquantel was used in its place as a more effective anti-tapeworm drug, however Niclosamide’s efficacy as an anti-cancer drug, including TNBC where very few options are effective, can not be overstated.30,31,32 Patient was dosed 1000mg daily, approximately 15mg/kg, below the average dose for tapeworms at 2000mg, and with 1 week of daily use results showed significant reduction of CTCs – 27% – from 26 to 19 per 7.5ml of blood.
Fig 4. 26 Circulating Tumor Cells per 7.5 ml
Fig 5. 19 Circulating Tumor Cells per 7.5ml
KI-67 expression by biopsy in contrast to the original tissue sample showed a similar reduction from 91-100% down to 65% during this same period of use. Additionally, Mutated TP53 gene was reduced from 31.6% to 16% in Blood Tumor Mutational Burden from 1.6m/MB to 0.5m/MB. Given CTCs failed to be inhibited via other methods of medication, including chemotherapy and immunotherapy, this is a very promising therapeutic option for TNBC patients, and cancer patients more broadly.33 Unfortunately Niclosamide use was prescribed and found to be effective at a terminal-stage in the cancer’s pathogenesis, weeks from diagnosis of lymphangitic carcinomatosis which has a 50% mortality rate of 3 months in one study.34
Fig 6. KI-67 Expression Reduced to 65%
Fig 7. Mutant TP53 at 31.6% in Blood Tumor Mutational Burden at 1.6m/MB
Fig 8. Mutant TP53 gene reduced to 16% in Blood Tumor Mutational Burden at 0.5m/MB
We can isolate the treatments’ positive and negative effects, and therefore any variables concluded, as they were prescribed and used at different times with no crossover of use by the patient. We can also conclude this wasn’t a unique case as the malignant behavior of the disease involved commonly elevated signal transduction pathways in TNBC and other cancers, while the protective oncogenes, inflammatory cytokines, PDL1 expression and immunogenicity were consistently evident with the preclinical molecular biology literature and clinical data.
METHODS
Tissue Exam: Estrogen Receptor (ER) and Progesterone Receptor (PR) assays were performed on 10% Neutral Buffered Formalin-fixed (NBF), paraffin-embedded tissue sections. The antibody vendor/clones are: Ventana ER (SP1) Rabbit Monoclonal and PR (1EZ) Rabbit monoclonal. The detection system used is Ventana iView DAB. Control slides containing known negative and positive tissue were used. Ki-67: Positive in 91-100% of malignant cells.
Mammogram Tomography: Computer aided detection used. Interpretation was made with the benefit of tomosynthesis imaging. Hand-held targeted breast ultrasound was also performed to contrast accuracy.
Viability Assays and Transcriptomic micro-Arrays: Isolation of the malignant cells using flow cytometry and negative selection (isolated 4.7 cells/7.5ml, SD +/- 0.3 cells). Isolated cells were expanded and split in two, from which one is utilized for viability assays and the other for transcriptomic micro-arrays.
Computed Tomography (CT) scan: Iodinated contrast agents were administered, standard coronal and sagittal reformats obtained on an independent work station for review via software delineation of tumor boundaries and rendered 3-dimensional structures.
Immunomagnetic separation (IMS): CTC enumeration was magnetically separated from the majority of other blood cells by using ferrofluid nanoparticles with antibodies that targeted epithelial cell adhesion molecules.
Oral-dosing Ivermectin: 36mg daily was initially given for 3 days, followed by 70mg daily for a period of approximately 16 weeks. Where tumor reduction recorded was succeeded by slowed tumor growth inhibition thereafter. However accurate records ended at 12 weeks when other treatments (which showed no response in CTCs and scans following) were attempted alongside and after Ivermectin doses ceased. Dosing was every morning, and recommended around 2hrs after a fat-rich meal to help with bioavailability. Dosage was chosen in an attempt to achieve an increased Cmax compared to standard dosing of 0.2mg/kg for parasites, with 2mg/kg clinically recorded as safely tolerated (Juarez, Mandy et al.). These higher concentrations are to reflect higher intracellular concentration within malignant cells. Further investigation is warranted of the potential benefit of higher-doses, including utilizing a 2mg/kg dose for cancer as Ivermectin’s efficacy in suppressing tumor growth is dose-dependent.
Oral-dosing Niclosamide: 1000mg daily for approximately 1 month with accurate records during this period, particularly the first 2 weeks of use. As 20mg/kg showed efficacy in Basal and TNBC lab reports (Londoño-Joshi, Angelina I. et al.), bioavailability was less an issue for maximizing efficacy or higher-doses for reaching Cmax potency, therefore no specific dietary regimen was prescribed. Dosing was also every morning.
COMPOUNDS AND MECHANISM OF ACTION
Niclosamide
An anthelmintic salicylamide derivative known to be an oxidative phosphorylation (OXPHOS) uncoupler and aerobic glycolysis inhibitor,35,36 similarly affecting cancer cells as the cells in parasitic worms. OXPHOS, and aerobic glycolysis in-particular are two of the more essential developments of tumorigenesis. This makes Niclosamide an inhibitor of CSCs directly, early in malignant development, and not just responsible for post-growth anti-tumor activity. Niclosamide’s ability to also downregulate multiple critical signal transduction pathways of many cancers is well researched, including with Basal-like Breast Cancers, which are up to 70-90% TNBC cases.37,38 Signal pathways Niclosamide inhibits include WNT/B-Catenin, PI3K/AKT, MAPK/MEK/ERK, mTOR, NF-KB, STAT3, and Notch.39,40 It should be noted the patient had elevated levels of PI3K, MEK-ERK, and mTOR, and many of these signal pathways are ubiquitous in the disease. WNT signalling is a master signal of CSCs and the tumor microenvironment (TME)41 and generator of cell growth, division and development.42 This patient case study, combined with other Basal TNBC reports, demonstrate Niclosamide’s efficacy as a potent TNBC and anti-cancer agent.43 Niclosamide’s broad-spectrum anti-cancer activity at standard dosing, while having no side effects therein, is remarkable. It was unfortunately discovered and used very late in this patient’s disease. This result could have had more profound outcomes earlier, and warrants further clinical trials and investigation of this FDA-Approved drug for cancer patients.
Ivermectin
A macrocyclic lactone from avermectins, made of actinomycetes cultures with the fungus Streptomyces, Ivermectin is a potent broad-spectrum antiparasitic which targets glutamate-gated chloride membranes.44 This, like Niclosamide, would explain in part Ivermectin’s mechanism of detection due to shared attributes with cancer cells, such as inducing apoptosis via chloride-dependent membrane hyperpolarization.45 Ivermectin has effective downregulation of numerous signal transduction pathways implicated in TNBC and a variety of cancers, such as PI3K, EGFR, HSP27, PAK1 (P21), mTOR, AKT, BCL-2, inflammasomes and cytokines such as NLRP3, HIF-1a, IL-1B, and TGF-b.46,47 It should be noted the patient had dysregulated EGF, HSP27, BCL-2, and TGF-b as well as cytokine increase as the disease progressed. Ivermectin’s ability to bind to the extracellular domain EGF receptor could explain its therapeutic potential in TNBC (especially with this patient’s overexpression of EGF) given the initial lack of hormonal receptors limiting treatment. Thus, it inhibits a signal cascade linked to further cell proliferation, such as ERK/AKT/NFKB.48 A general view of Ivermectin’s anti-tumor activity is as follows: acts as an ionophore and up-regulates chloride channels to induce apoptosis. Downregulating the function of mitochondrial-complex-I, ivermectin inhibits the electronic oxidative phosphorylation pathway that activates oxygen consumption rate (OCR) to generate ATP energy for the cell. Ivermectin induces Immunogenic Cell Death (ICD) through the stimulation of an ATP and HMGB1 enriched Tumor Microenvironment (TME).49 These inflammatory pathways can reduce the efficacy of ICD and therefore Anti-PD1 immunotherapies such as pembrolizumab due to the upregulation of JAK 1 and/or JAK 2 STAT1 pathway.50 The ICD capacity of Ivermectin also helps recruit CD4/CD8 T-Cells, which is what made it an effective combination with pembrolizumab in fully curing TNBC animal models. Ivermectin’s induction of ICD is partly seen in TNBC through the lens of allosterically potentiated P2X4 and P2X7 receptors, and caspase-1/Interleukin-1 (IL-1) through stimulation of the ATP-enriched TME.
Berberine
Berberine is an isoquinoline alkaloid present in Berberis, Hydrastis canadensis and Coptidis rhizoma. Research suggests glutamate from cancer cells activates–or is dysregulated by–Ca2+-permeable AMPARs and NMDARs which causes migration and invasion. Ca2+ influx can cause excitotoxic death of cells for TME invasion.51 Berberine inhibits glutamate release by a reduction of Ca2+ influx through Cav2.1 channels.52 Ca2+ signaling is also involved in neurological disease such as Alzheimer’s and Autism, and Ca2+-activated transcription factors regulate the recruitment of chromatin remodeling complexes into their target genes, and Ca2+-sensing proteins modulate their activity and intracellular localization.53 This makes way for additional downregulation of key cancer signalling and energy ATP production via modulating Glutamate and Ca2+, with potential epigenetic modification boosted significantly alongside controlled-fasting and Ivermectin’s aforementioned mechanism of action. Berberine is studied extensively for its anti-cancer effects.54
EGCG
Epigallocatechin gallate (EGCG), a natural polyphenol extracted from green tea, has been significantly reviewed for its anti-cancer effects.55 EGCG’s mechanism of action includes various biomolecular targets; STAT, NF-κB, TGF-β, TLR4, and PI3K/Akt. Perhaps most notably, EGCG downregulates PDL1, this Anti-PDL1 effect, which is a tumor cell blockade binding to PD1 on Natural Killer T-Cells (CD4/CD8) inhibiting their ability to penetrate malignant cells and induce ICD. EGCG mirrors Anti-PD1 immunotherapies such as pembrolizumab but acting on the tumor cell instead of the T-Cell. This could explain synergy with Ivermectin’s ICD effects and has potential to be furthered synergistically with pembrolizumab (synergy would also be found with Niclosamide but wasn’t used at the time of dosing).56 It could also reduce autoimmune symptoms from immunotherapies by reducing the amount needed for effectively enhancing CD4/CD8 T-Cell infiltration activity.57
Vitamin D
Shown to have a cytoprotective role in TNBC, vitamin D (25(OH)D) in part could reverse TP53 degradation, which was significant for the patient and present in many cancers, as well as decreased Epithelial–Mesenchymal Transition (EMT) cancer cell stemness, consequence of increased CD44 expression plus cytokines IL-6 and IL-8.58
EPIGENETIC REPROGRAMMING
Patient was diagnosed via tissue exam as a typical TNBC phenotype with no estrogen receptor (ER), progesterone receptor (PR), or human epidermal growth factor receptor 2 (HER-2) expression. With 26 days between original tissue sample and viability assays & transcriptomic micro-Arrays data, we see a clear cellular phenotype epigenetic alteration with 25% ER and 15% PR. Avermectins have been previously noted to epigenetically modify TNBC cell lines in metastatic mice and human cell lines with re-expression of ESR1 via SIN3-PAH2 protein transcription factors.59 However, we provide the first human patient case study of this epigenetic restoration, with PGR expression as well. The same complex SIN3-PAH2-MAD1 was also responsible for RARB expression in breast cancer models. As we discussed previously, functionally reprogramming inactive genes in a human being safely appears to require a more systemic approach than one target area or drug, however it is additional data to point to our results being consistent and not an aberration. As an example, mTOR signaling is required for ESR1 activity, and if mTOR is inhibited, ESR1 will remain inactivated, but mTOR also drives malignancy, so only a systemic gene reprogramming approach will be sufficient for human patients.
SIN3 protein is a transcriptional regulator that functions as the central scaffold unit of the multi-protein SIN3/HDAC co-repressor complex.60 Histone deacetylases (HDACs) are enzymes that remove acetyl groupings from proteins, which alters how histones bind to DNA. Acetylating the histone reduces how tightly DNA chromatin is wrapped around nucleosomes, opening transcription to affect gene expression. Methylation happens to the bases of DNA itself and "silences" it, reducing gene transcription. HDACs in this case typically tighten DNA chromatin structures (a core of these repeating units are a nucleosome) to decrease gene expression. HDACs are highly relevant in TNBC and other cancers, with significance in the pathogenesis of Autism.61,62
Some epigenetic alterations could also be Ivermectin’s activation of AMPK as well as its interaction via mTOR, which are known epigenetic modulators.63,64,65 AMPK and mTOR converge in a variety of factors including acetylation or methylation, chromatin remodelling and epigenetic enzymes for transcription events. Berberine also supports AMPK activation, as does EGCG.66,67 AMPK activation can inhibit HDAC5 and modulate HDACs more broadly, with HDAC inhibitors reported to have re-established estrogen receptor expression.68,69,70
1980s HeLa cells experiments indicated phosphorylation of histone H3 as Ca2+-dependent, with HDACi sodium butyrate pretreatment potentiating Ca2+-induced phosphorylation71. Berberine is evidenced to affect Ca2+ via intracellular levels and oscillation, and HDACs via epigenetic chromatin remodelling72. Berberine-treated cells in one study showed upregulation of histone acetyltransferase CREBBP and EP300, essential for cell differentiation & development, cell cycles, and DNA repair, and playing a role in epigenetic regulation by adding acetyl groups to histone proteins affecting gene expression by making chromatin more accessible for transcription with many signalling factors and receptors.73,74 There is also evidence to suggest Berberine induces acetylation of a-tubulin, a microtubule building block affecting protein isotype expression.75 In addition to this Ca2+ epigenetic process, we see research point to aberrant calcium signaling downstream mutations in TP53, PI3K, and increased therapy resistance in TNBC.76
EGCG was reported to suppress DNA methyltransferase, leading to cytosine-phosphate-guanine de-methylation and to restore silenced tumor-suppressor genes.77 Evidence suggests EGCG also alters histone acetylation (H3K9 and H3ac), as well as methylation of H3K4me3 and H3K9me3 chromatin marks. We see Histones more systemically in bioscientific analysis playing a key role in estrogen and progesterone receptors, with more global impact upon other epigenetic mechanisms.78,79
It should be repeated that these results, i.e., ESR1 being active, was underexpressed after the controlled-fasting ended, demonstrating limited epigenetic use in TNBC specifically without a more potent metabolic reprogramming to accompany the genetic restoration.
The Critical PI3K-mTOR Oncogenic Bypass
Patient had overexpression of PIK3CA in the same RNA liquid biopsy sample that demonstrated active (but then underexpressed) ESR1. By inhibiting PI3K, we removed the negative regulator that keeps mTOR locked in its cancer-survival mode. This allows mTOR-S6 phosphorylation to function epigenetically rather than oncogenically, facilitating demethylation and ESR1/PGR reactivation. The treatment exploited a favorable biochemical window for coordinated metabolic, epigenetic, and transcriptional changes: not just shutting down an oncogenic pathway, but redirecting it to achieve controlled epigenetic repair within favored metabolic conditions (a-KG not Glycolysis), rather than conditions and signaling that favor tumor survival, is what made this safe & effective.
Streptomyces-derived compounds (including avermectins) are known PI3K inhibitors. PI3K normally activates AKT, which in turn activates mTORC1. In cancer (including TNBC), PI3K-AKT-mTOR signaling is often hyperactive, maintaining proliferation, stemness (CSCs), and survival.
This rebalancing was critical for epigenetic re-expression of ESR1 and PGR and marks a departure from prior mTOR-inhibition strategies, which often stall both tumor and recovery signals.80 mTOR-S6 Phosphorylation under these conditions became epigenetic, not oncogenic. Without PI3K's aggressive input, mTOR-S6 could serve in its basal, chromatin-regulatory role, assisting with histone modification, especially promoting H3K27me3 demethylation and ESR1/PGR re-expression.81 Effectively turning a typically cancer-promoting pathway into a recovery-enabling one by restraining the upstream driver (PI3K).
Selective metabolic and ion flux conditions with the ATP/Glycolysis⁻flux and switch to a-KG in TNBC likely further contributed to modulating the ionic environment for epigenetic writers and erasers, given the active but underexpression of ESR1 post-fasting. SIN3-HDAC inhibition plus mTOR-S6 assistance became a recovery axis rather than a tumor-promoting one.
We modulated the metabolic environment to suppress oncogenic signaling and support histone remodeling for gene reactivation. Inhibiting mTOR/S6 directly would shut down both the malignant proliferative signals and the epigenetic modifications needed for gene re-expression. That’s why standard mTOR inhibitors fail to produce durable epigenetic recovery in cancers, or why epigenetic modifications of genetic re-expression (such as those in lab studies) are high-risk to facilitate results in human patients, as mTOR left unchecked to facilitate epigenetics could further proliferation or dysregulated signaling.
First-of-Its-Kind Human Evidence – we have successfully demonstrated epigenetic reprogramming and re-expression of undetectable, silenced genes and receptors in a living human, not just upregulation (modulation) of expression. Which has never been documented in medical literature before.
Genetic Markers Confirm Modification – our data shows clear changes in gene expression, receptor reactivation (ESR1/PGR), and histone remodeling (HDAC-dipeptide increase, DNA methyltransferase reduction), proving functional epigenetic reprogramming.
Functional Evidence of Tumor Reduction – The correlation of tumor reduction and receptor re-expression strongly supports that treatment not only impacted gene expression but also had a direct therapeutic effect.
Metabolic & Molecular Pathway Validation – our work connects metabolic and mitochondrial complexes, ATP flux, and PI3K inhibition with histone acetylation and gene activation—linking metabolism directly to epigenetics in a way not previously demonstrated in a human subject.
Multi-Disease Potential – our approach operates at the fundamental biochemical level (metabolic reprogramming, histone remodeling, nucleotide recruitment), this methodology is applicable to many genetic diseases, not just cancer.
Using logistic analysis on epigenetic profiles, medications used and their relevant signal pathways, especially upregulated in TNBC, we can elucidate how the epigenetics in a human patient have occurred with this treatment. Much more research needs to be carried out before any conclusions for achieving this on a repeatable, effective basis can be reached.
Fig 9. ESR1/PGR Activated from Previously Silenced: Estrogen Receptor 25%, Progesterone Receptor 15%
Fig 10. Ribonucleotide Reductase (RNR) converts ribonucleotides into deoxyribonucleotides, essential for DNA synthesis. An increase to 25% suggests active DNA repair and replication.
RESULTS
Gene Activation and Receptor Re-Expression:
Quantitative increases in ESR1 (ERa 25%) and PGR (PR 15%) from undetectable and <1% respectively.
Molecular Marker Analysis:
mTOR-S6 stabilization (25%), DNMT (-20%), HDAC-dipeptide expression (40%), and increased ribonucleotide reductase (25%). These collectively demonstrate active genetic reprogramming, expression, demethylation and repair.
Tumor Reduction:
Detailed measurements and volumetric analysis showing 68–77% reduction.
The Gene Activation, Molecular Sample and Tumor reduction were within 1 month of treatment.
CSC/CTC Marker Reduction:
Decreased tumor cell stemness and metastatic potential at 27% reduction.
Tumor Mutational Burden, TP53 Reduction:
Mutated TP53 gene reduced from 31.6% to 16% in Blood Tumor Mutational Burden from 1.6m/MB to 0.5m/MB.
Proliferation marker (KI-67):
Reduced from 91–100% to 65%
The proliferation and mutational reduction was with 1-2 weeks use.
HDACs are key regulators of chromatin structure—they remove acetyl groups from histones, tightening DNA and repressing gene expression. A 40% expression of HDAC-dipeptide shows active histone remodeling via HDAC inhibition, balancing acetylation and deacetylation dynamics.
DNA methyltransferases (DNMTs) are enzymes that catalyze the addition of methyl groups to cytosine bases in DNA, methylation plays a crucial role in regulating and repressing gene expression and also maintaining genome stability. DNMTs at -20% shows active DNA demethylation for altering gene expression.
Ribonucleotide Reductase (RNR) converts ribonucleotides into deoxyribonucleotides, essential for DNA synthesis. An increase to 25% suggests active DNA repair and replication, further proving epigenetic modification was sustained and not just transient. This aligns with our model and evidence that chromatin was loosened for transcriptional activation and cellular reprogramming. RNR is a critical enzyme for DNA synthesis and repair. Its function ties directly into the epigenetic remodeling process.The fact that RNR was elevated but not excessive suggests controlled DNA synthesis, supporting the genetic re-expression rather than oncogenic mutation accumulation.
mTOR at 25% confirms it was modulated rather than suppressed, as mTOR is a key phosphorylation of systemic epigenetic pathways that play a part in cell size, function, metabolism, and immune response. mTOR was stabilized, not inhibited, allowing histone modification without triggering oncogenic proliferation.
These markers show how treatment successfully modified histone regulation to reactivate ESR1/PGR. The detected HDAC activity suggests an ongoing epigenetic shift, rather than a complete reversal. This supports that our method actively remodels chromatin rather than just transiently activating genes and is therefore epigenetically reprogrammed, not just temporarily expressed.
Ivermectin dosage of 70mg daily at around 1.1mg/kg with CT scan 20 days post-mammogram showing a tumor reduction of approx. 70%. 0.6cm growth weekly growth inhibition thereafter while proliferation continued (approx. 1cm down to 0.4cm).
Ivermectin, Berberine, EGCG, Vitamin D and controlled-fasting use in this same period produced TNBC cells with hormonal receptors from originally undetectable to 25% in Estrogen, and <1% to 15% in Progesterone. Symptom patterns (neural hyperactivity and eyesight sensitivity) are consistent with RARB re-expression in previous animal models demonstrating it via the exact mechanism of action as ESR1/ERa expression.
Niclosamide was dosed at 1000mg daily, approximately 15mg/kg, below the average dose for tapeworms at 2000mg, and with 1-2 weeks of daily use results showed significant reduction of CTCs at around 27% from 26 to 19 per 7.5ml of blood. KI-67 expression by biopsy in contrast to the original tissue sample showed a similar reduction from 91-100% down to 65%, and Mutant TP53 down from 31.6% to 16% during this same period of use.
Consideration of measurement discrepancies (ultrasound vs. CT). A reasonable discrepancy range for ultrasound vs. CT could be 5-15% overestimation in ultrasound measurements. Applying this to the initial tumor size (5.6 × 3.8 × 4.2 cm):
If ultrasound overestimated by 10%, the actual initial tumor size could have been closer to 5.0 × 3.4 × 3.8 cm.
This would make the total volume around 64.6 cm³ instead of 85.4 cm³.
Comparing to the post-treatment CT scan (4.1 × 2.0 × 2.5 cm = 20.5 cm³):
Best-case scenario (original measurements accurate): 77.06% reduction.
Worst-case scenario (10% initial overestimation): 68.3% reduction (from 64.6 cm³ to 20.5 cm³).
Thus, even in a worst-case discrepancy scenario, tumor reduction would still be around 68%, which remains a highly significant response to treatment, hence we have an approximately 70% reduction throughout this paper.
The initial protocol’s growth inhibition (1cm to 0.4cm) is estimated based upon the original speed of growth, and following growth between scans. This measurement is therefore not as directly tracked as the clinically-reviewed tumor reductions and should be studied further for more accuracy.
As mTOR can further malignant proliferation instead of epigenetic modification, this approach must be further replicated, fine-tuned and perfected for safety before being deployed in human patients.
DISCUSSION
The First Human Demonstration of Functional Epigenetic Reprogramming & Restoration of Silenced Genes
Achieved in a living human with measurable genetic and phenotypic changes, ESR1, PGR gene activation with ER/PR receptor re-expression. This is an experimental first, not just an extension of existing work.
Revealed a new Multi-System Mechanism:
Glycolysis-Glutaminolysis → ATP Flux and Alpha-Ketoglutarate switch → PI3K oncogenic bypass for mTOR signaling → SIN3-HDAC Inhibition → HDAC-Dipeptide, RNR, DNMT’s and mTOR recruitment (Histone Repression Loss and Active Repair) → Gene Re-expression.
This bridges oncology, metabolism, and gene expression in a way no previous human treatment paper has shown.
Reversal of Malignant Signaling to Promote Genetic Repair:
By selectively inhibiting PI3K while retaining mTOR-S6, we did what most therapies don’t;
stopped cancer without stopping the body’s ability to repair itself from genetic silencing.
Clinical Impact:
Tumor Shrinkage, Receptor Expression, Genetic Activation. A 68–77% tumor reduction, with genetic evidence, receptor re-expression, and biochemical stability—without toxicity or gene editing—has never been done in a human being safely. There are also extremely limited examples of CTC/CSC/KI-67 and TP53 reductions in humans without toxicity.
There is much to gain by repurposing FDA approved drugs with a good safety profile for use in TNBC and cancers more widely difficult to treat with conventional methods. This represents the first publicly documented case of multi-locus gene reprogramming and chromosome loci restoration in a human being without genetic editing, and while the exact mechanism of action remains to be studied, paves the way for more breakthroughs in genetic disease medicine via metabolic and epigenetic reprogramming.
Future Research
Over 1 billion adults and children live with epigenetic dysregulation and gene silencing—underlying drivers of cancer, neurodegeneration, autoimmune conditions, autism, and cognitive disorders. This case study utilized just two primary pharmacological interventions alongside three plant-derived compounds, yet achieved systemic gene reprogramming and reversal of aggressive cancer progression with zero observed toxicity.
We have since identified additional synergistic compounds capable of inducing immunogenic cell death (ICD), downregulating oncogenic self-renewal pathways in the tumor microenvironment (TME), and sustaining metabolic and transcriptional remodeling. These agents target multiple axes: cancer stem cell (CSC) inhibition, nutrient stress amplification, and epigenetic repair. Future versions of this protocol aim to balance efficacy with prevention of immune overactivation, autoimmunity, or recurrence.
Our long-term objective is to prevent, not just treat, cancer—by reprogramming tumor suppressor genes before malignancy takes hold.
Beyond oncology, this approach may hold promise for neurodevelopmental and cognitive disorders, such as autism, where histone repression and epigenetic instability drive lifelong disability. By reversing maladaptive gene silencing safely and non-invasively, this methodology could enable a new era of human-first epigenetic therapeutics.
This publication marks the beginning. Our goal now is to validate, refine, and extend this work into rigorous translational research—honoring the breakthrough that came at great personal cost by ensuring it serves the millions still waiting for hope.
References
[1] Won, Kwang-Ai, Spruck Charles, Triple-negative breast cancer therapy: Current and future perspectives (Review), International Journal of Oncology (December 2020) Volume 57 Issue 6, Print ISSN: 1019-6439, Online ISSN:1791-2423 https://doi.org/10.3892/ijo.2020.5135
[2] Almansour, Nahlah Makki, Triple-Negative Breast Cancer: A Brief Review About Epidemiology, Risk Factors, Signaling Pathways, Treatment and Role of Artificial Intelligence, Frontiers in Molecular Biosciences, Sec. Molecular Diagnostics and Therapeutics Volume 9 (2022) https://doi.org/10.3389/fmolb.2022.836417
[3] Qayoom, Hina et al. An Insight Into the Cancer Stem Cell Survival Pathways Involved in Chemoresistance in Triple-Negative Breast Cancer, Future Oncology Volume 17 (2021) https://doi.org/10.2217/fon-2021-0172
[4] He, Lin et al. The Role of Breast Cancer Stem Cells in Chemoresistance and Metastasis in Triple-Negative Breast Cancer, Cancers Volume 13, Issue 24 (2021) https://doi.org/10.3390/cancers13246209
[5] Icard Philippe, Lincet Hubert, The reduced concentration of citrate in cancer cells: An indicator of cancer aggressiveness and a possible therapeutic target, Drug Resistance Updates, Volume 29 (2016)
https://doi.org/10.1016/j.drup.2016.09.003
[6] A.M. Li, B. He, D. Karagiannis, Y. Li, H. Jiang, P. Srinivasan, Y. Ramirez, M. Zhou, C. Curtis, J.J. Gruber, C. Lu, E.B. Rankin, & J. Ye, Serine starvation silences estrogen receptor signaling through histone hypoacetylation, Proc. Natl. Acad. Sci. U.S.A. 120 (38) e2302489120, https://doi.org/10.1073/pnas.2302489120 (2023)
[7] Verma, A., Singh, A., Singh, M.P. et al. EZH2-H3K27me3 mediated KRT14 upregulation promotes TNBC peritoneal metastasis. Nat Commun 13, 7344 (2022). https://doi.org/10.1038/s41467-022-35059-x
[8] Dahiya, Nisha & Leibovitch, Boris & Kadamb, Rama & Bansal, Nidhi & Waxman, Samuel. (2022). The Sin3A/MAD1 Complex, through Its PAH2 Domain, Acts as a Second Repressor of Retinoic Acid Receptor Beta Expression in Breast Cancer Cells. Cells. 11. 1179. 10.3390/cells11071179
[9] Interference with Sin3 function induces epigenetic reprogramming and differentiation in breast cancer cells. Crossref DOI link: https://doi.org/10.1073/pnas.1006737107 Published Online: 2010-06-14
[10] Chung C, et al. Integrated Metabolic and Epigenomic Reprograming by H3K27M Mutations in Diffuse Intrinsic Pontine Gliomas. Cancer Cell. 2020 Sep 14;38(3):334-349.e9. doi:10.1016/j.ccell.2020.07.008
[11] Ma Z, Wang H, et al. Epigenetic drift of H3K27me3 in aging links glycolysis to healthy longevity in Drosophila. Elife. 2018 May 29;7:e35368. doi:10.7554/eLife.35368
[12] Fei Huang et al. ,Control of histone demethylation by nuclear-localized α-ketoglutarate dehydrogenase. Science 381, eadf8822 (2023). DOI:10.1126/science.adf8822
[13] Sungmin Joo, et al. α-ketoglutarate suppresses immediate early gene expression in cancer cells, Biochemical and Biophysical Research Communications, Volume 637, 2022, https://doi.org/10.1016/j.bbrc.2022.11.021.
[14] Jiang J, Srivastava S, Zhang J. Starve Cancer Cells of Glutamine: Break the Spell or Make a Hungry Monster? Cancers (Basel). 2019 Jun 11;11(6):804. doi:10.3390/cancers11060804
[15] Nichol JN, et al. H3K27 Methylation: A Focal Point of Epigenetic Deregulation in Cancer. Adv Cancer Res. 2016;131:59-95. doi:10.1016/bs.acr.2016.05.001. (2016)
[16] Xiao Mengtao, et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors Published in Advance June 7, 2012, doi:10.1101/gad.191056.112 Genes & Dev. (2012)
[17] Beacon, T.H., Delcuve, G.P., López, C. et al. The dynamic broad epigenetic (H3K4me3, H3K27ac) domain as a mark of essential genes. Clin Epigenet 13, 138 (2021). https://doi.org/10.1186/s13148-021-01126-1
[18] Fendt, Sarah-Maria et al. Metformin Decreases Glucose Oxidation and Increases the Dependency of Prostate Cancer Cells on Reductive Glutamine Metabolism, American Association for Cancer Research Volume 73, Issue 14 (2013) https://doi.org/10.1158/0008-5472.CAN-13-0080
[19] Elayapillai, Suganthrapriya et al. Potential and mechanism of mebendazole for treatment and maintenance of ovarian cancer, Gynecologic Oncology Volume 160, Issue 1 P302-311 (2021) https://www.gynecologiconcology-online.net/article/S0090-8258(20)34018-X/
[20] Nguyen, Jolie et al. Oral Fenbendazole for Cancer Therapy in Humans and Animals, Anticancer Research Volume 44, Issue 9 (2024) https://doi.org/10.21873/anticanres.17197
[21] Kelkel, Mareike et al. Potential of the Dietary Antioxidants Resveratrol and Curcumin in Prevention and Treatment of Hematologic Malignancies, Molecules Volume 15, Issue 10 (2010) https://doi.org/10.3390/molecules15107035
[22] Kantley, Lewis Intravenous High-Dose Vitamin C in Cancer Therapy, National Cancer Institute, The RAS Initiative (2024) https://www.cancer.gov/research/key-initiatives/ras/news-events/dialogue-blog/2020/yun-cantley-vitamin-c
[23] Kuršvietienė, Lolita Selenium Anticancer Properties and Impact on Cellular Redox Status Antioxidants Volume 9, Issue 1 (2020) https://doi.org/10.3390/antiox9010080
[24] Draganov, Dobrin et al. Ivermectin converts cold tumors hot and synergizes with immune checkpoint blockade for treatment of breast cancer, Nature NPJ Breast Cancer 7, Article number: 22 (2021) https://doi.org/10.1038/s41523-021-00229-5
[25] Tang, Mingyang et al. Ivermectin, a potential anticancer drug derived from an antiparasitic drug, Elsevier Pharmacological Research Volume 163 (2021) https://doi.org/10.1016/j.phrs.2020.105207
[26] Nambara, Sho et al. Antitumor effects of the antiparasitic agent ivermectin via inhibition of Yes-associated protein 1 expression in gastric cancer, Oncotarget Volume 8, Issue 64 (2017) https://doi.org/10.18632/oncotarget.22587
[27] Bhumsuk, Keam et al. Ki-67 can be used for further classification of triple negative breast cancer into two subtypes with different response and prognosis, Breast Cancer Research volume 13, Article number: R22 (2011) https://doi.org/10.1186/bcr2834
[28] Rais, Ghizlane et al. Assessment of the Predictive Role of Ki-67 in Breast Cancer Patients’ Responses to Neoadjuvant Chemotherapy, European Journal of Breast Health 2024;20(3):199-206 https://doi.org/10.4274/ejbh.galenos.2024.2024-3-8
[29] Srivastara, Pooja et al. Clinical-pathologic characteristics and response to neoadjuvant chemotherapy in triple-negative low Ki-67 proliferation (TNLP) breast cancers, Nature NPJ Breast Cancer 8, Article number: 51 (2022) https://doi.org/10.1038/s41523-022-00415-z
[30] Chen, Wei et al. Niclosamide: Beyond an antihelminthic drug, Elsevier Cellular Signalling Volume 41 (2018) https://doi.org/10.1016/j.cellsig.2017.04.001
[31] J.H. Kim, et al. dual-action niclosamide-based prodrug that targets cancer stem cells and inhibits TNBC metastasis, Proc. Natl. Acad. Sci. U.S.A. 120 (21) e2304081120, doi.org/10.1073/pnas.2304081120
[32] Clinical Care of Dipylidium CDC, Praziquantel, Niclosamide (2024) https://www.cdc.gov/dipylidium/hcp/clinical-care/?CDC_AAref_Val=https://www.cdc.gov/parasites/dipylidium/health_professionals/index.html
[33] Wang, LH., Xu, M., Fu, LQ. et al. The Antihelminthic Niclosamide Inhibits Cancer Stemness, Extracellular Matrix Remodeling, and Metastasis through Dysregulation of the Nuclear β-catenin/c-Myc axis in OSCC. Sci Rep 8, 12776 (2018). https://doi.org/10.1038/s41598-018-30692-3
[34] Toshima H, et al. Two cases of lymphangitic carcinomatosis as the primary symptom of colorectal carcinoma that achieved complete remission using combination anti-EGFR antibody therapy. Onco Targets Ther. 2019;12:2089-2093 https://doi.org/10.2147/OTT.S194224
[35] Kumar, R., Coronel, L., Somalanka, B. et al. Mitochondrial uncoupling reveals a novel therapeutic opportunity for p53-defective cancers. Nat Commun 9, 3931 (2018). https://doi.org/10.1038/s41467-018-05805-1
[36] Shangguan, F., Liu, Y., Ma, L., Qu, G., Lv, Q., An, J., Yang, S., Lu, B., Cao, Q. (2020). Niclosamide inhibits ovarian carcinoma growth by interrupting cellular bioenergetics. Journal of Cancer, 11(12), 3454-3466. https://doi.org/10.7150/jca.41418.
[37] Londoño-Joshi, Angelina I. et al. Mol Cancer Ther (2014) 13 (4): 800–811 https://doi.org/10.1158/1535-7163.MCT-13-0555
[38] Badowska-Kozakiewicz AM, Budzik MP. Immunohistochemical characteristics of basal-like breast cancer. Contemporary Oncology (2016) 20(6):436-443. doi.org/10.5114/wo.2016.56938.
[39] Arend R. C., et al. Niclosamide and its analogs are potent inhibitors of Wnt/β-catenin, mTOR and STAT3 signaling in ovarian cancer. Oncotarget. (2016) 7: 86803-86815. https://doi.org/10.18632/oncotarget.13466
[40] Cheng B, Morales LD, Zhang Y, Mito S, Tsin A Niclosamide induces protein ubiquitination and inhibits multiple pro-survival signaling pathways in the human glioblastoma U-87 MG cell line, PLoS ONE (2017) 12(9): e0184324. https://doi.org/10.1371/journal.pone.0184324
[41] Cristina Castañeda-Patlán, M., Fuentes-García, G., & Robles-Flores, M. Wnt Signaling as a Master Regulator of Immune Tolerance in a Tumor Microenvironment, IntechOpen 2019) doi.org/10.5772/intechopen.81619
[42] Qin, Kevin et al. Canonical and noncanonical Wnt signaling: Multilayered mediators, signaling mechanisms and major signaling crosstalk, Genes & Diseases (2024) Volume 11, Issue 1 doi.org/10.1016/j.gendis.2023.01.030
[43] Haowen, Jiang et al. The magic bullet: Niclosamide, Frontiers Oncology Sec. Cancer Metabolism Volume 12 (2022) doi.org/10.3389/fonc.2022.1004978
[44] Atif M, Estrada-Mondragon A, et al. Effects of glutamate and ivermectin on single glutamate-gated chloride channels of the parasitic nematode H. contortus, PLOS Pathogens (2017) 13(10): e1006663. https://doi.org/10.1371/journal.ppat.1006663
[45] Sumaiya Sharmeen et al. The antiparasitic agent ivermectin induces chloride-dependent membrane hyperpolarization and cell death in leukemia cells, Blood (2010) 116 (18): 3593–3603. doi: https://doi.org/10.1182/blood-2010-01-262675
[46] Mingyang, Tang, et al. Ivermectin, a potential anticancer drug derived from an antiparasitic drug, Pharmacological Research Volume 163, (2021) https://doi.org/10.1016/j.phrs.2020.105207.
[47] Abd-Elmawla, M.A., Ghaiad, H.R., Gad, E.S. et al. Suppression of NLRP3 inflammasome by ivermectin ameliorates bleomycin-induced pulmonary fibrosis. J. Zhejiang Univ. Sci. B (2023) 24, 723–733 https://doi.org/10.1631/jzus.B2200385
[48] Jiang, L., Wang, P., Sun, YJ. et al. Ivermectin reverses the drug resistance in cancer cells through EGFR/ERK/Akt/NF-κB pathway. J Exp Clin Cancer Res (2019) 38, 265 https://doi.org/10.1186/s13046-019-1251-7
[49] Juarez M, Schcolnik-Cabrera A, Dueñas-Gonzalez A. The multitargeted drug ivermectin: from an antiparasitic agent to a repositioned cancer drug. Am J Cancer Res. (2018) 1;8(2):317-331. PMID: 29511601; PMCID: PMC5835698. https://pmc.ncbi.nlm.nih.gov/articles/PMC5835698/
[50] Luo, N., Formisano, L., Gonzalez-Ericsson, P. I., et al. Melanoma response to anti-PD-L1 immunotherapy requires JAK1 signaling, but not JAK2. OncoImmunology (2018) 7(6). https://doi.org/10.1080/2162402X.2018.1438106
[51] Jae-son, So et al. Mechanisms of Invasion in Glioblastoma: Extracellular Matrix, Ca2+ Signaling, and Glutamate, Frontiers in Cellular Neuroscience Volume 15 (2021) https://doi.org/10.3389/fncel.2021.663092
[52] Lin TY, Lin YW, et al. Berberine Inhibits the Release of Glutamate in Nerve Terminals from Rat Cerebral Cortex. PLOS ONE (2013) 8(6): e67215. https://doi.org/10.1371/journal.pone.0067215
[53] Andrés Hernández-Oliveras, Angel Zarain-Herzberg, The role of Ca2+-signaling in the regulation of epigenetic mechanisms, Cell Calcium Volume 117 (2024) https://doi.org/10.1016/j.ceca.2023.102836.
[54] Almatroodi SA, Alsahli MA, Rahmani AH. Berberine: An Important Emphasis on Its Anticancer Effects through Modulation of Various Cell Signaling Pathways, Molecules (2022) 27(18):5889. https://doi.org/10.3390/molecules27185889
[55] Li, Dongming et al. The roles of epigallocatechin gallate in the tumor microenvironment, metabolic reprogramming, and immunotherapy, Frontiers in Immunology Volume 15 (2024) https://doi.org/10.3389/fimmu.2024.1331641
[56] Luo, F., Luo, M., Rong, QX. et al. Niclosamide, an antihelmintic drug, enhances efficacy of PD-1/PD-L1 immune checkpoint blockade in non-small cell lung cancer. j. immunotherapy cancer 7, 245 (2019). https://doi.org/10.1186/s40425-019-0733-7
[57] Autoimmune complications of immunotherapy, New Zealand Medicines and Medical Devices Safety Authority (2023) https://medsafe.govt.nz/profs/PUArticles/June2023/Autoimmune-complications-immunotherapy.html
[58] Blasiak J, Pawlowska E, Chojnacki J, Szczepanska J, Fila M, Chojnacki C. Vitamin D in Triple-Negative and BRCA1-Deficient Breast Cancer—Implications for Pathogenesis and Therapy, International Journal of Molecular Sciences (2020) 21(10):3670. https://doi.org/10.3390/ijms21103670
[59] Yeon-Jin Kwon, et al. Selective Inhibition of SIN3 Corepressor with Avermectins as a Novel Therapeutic Strategy in Triple-Negative Breast Cancer, Mol Cancer Ther 1 (2015) 14 (8): 1824–1836. https://doi.org/10.1158/1535-7163.MCT-14-0980-T
[60] Konwar, C., Maini, J., Kohli, S. et al. SIN-3 functions through multi-protein interaction to regulate apoptosis, autophagy, and longevity in Caenorhabditis elegans, Sci Rep 12, 10560 (2022). https://doi.org/10.1038/s41598-022-13864-0
[61] Ropero Santiago, Esteller Manel, The role of histone deacetylases (HDACs) in human cancer, Molecular Oncology, 1, (2007) doi.org/10.1016/j.molonc.2007.01.001.
[62] Tseng, Chieh-En Jane et al. Epigenetics of Autism Spectrum Disorder: Histone Deacetylases, Biological Psychiatry, Volume 91, Issue 11, 922 - 933 https://www.biologicalpsychiatryjournal.com/article/S0006-3223(21)01832-1/abstract
[63] Wang X, Wang J, et al. Cytotoxicity and Autophagy Induced by Ivermectin via AMPK/mTOR Signaling Pathway in RAW264.7 Cells, Molecules (2023) 28(5):2201. https://doi.org/10.3390/molecules28052201
[64] Gongol B, Sari I, et al. AMPK: An Epigenetic Landscape Modulator, International Journal of Molecular Sciences (2018) 19(10):3238. https://doi.org/10.3390/ijms19103238
[65] Zhao T, Fan J, Abu-Zaid A, et al. Nuclear mTOR Signaling Orchestrates Transcriptional Programs Underlying Cellular Growth and Metabolism, Cells (2024) 13(9):781. https://doi.org/10.3390/cells13090781
[66] Yun S. Lee, Woo S. Kim, et al. Berberine, a Natural Plant Product, Activates AMP-Activated Protein Kinase With Beneficial Metabolic Effects in Diabetic and Insulin-Resistant States, Diabetes (2006) 55 (8): 2256–2264. https://doi.org/10.2337/db06-0006
[67] Shirin Pournourmohammadi, Mariagrazia Grimaldi, et al.
Epigallocatechin-3-gallate (EGCG) activates AMPK through the inhibition of glutamate dehydrogenase in muscle and pancreatic ß-cells: A potential beneficial effect in the pre-diabetic state? The International Journal of Biochemistry & Cell Biology, Volume 88 (2017) https://doi.org/10.1016/j.biocel.2017.01.012.
[68] Ke, R., Xu, Q., et al. Mechanisms of AMPK in the maintenance of ATP balance during energy metabolism, Cell Biol Int (2018) 42: 384-392. https://doi.org/10.1002/cbin.10915
[69] Li, Z., Zhu, W.G. et al. Targeting Histone Deacetylases for Cancer Therapy: From Molecular Mechanisms to Clinical Implications, International Journal of Biological Sciences (2014) 10(7), 757-770. https://doi.org/10.7150/ijbs.9067.
[70] Scott Thomas, Pamela N. Munster, Histone deacetylase inhibitor induced modulation of anti-estrogen therapy, Cancer Letters Volume 280, Issue 2 (2009) https://doi.org/10.1016/j.canlet.2008.12.026.
[71] Andrés Hernández-Oliveras, Angel Zarain-Herzberg, The role of Ca2+-signaling in the regulation of epigenetic mechanisms, Cell Calcium Volume 117 (2024) https://doi.org/10.1016/j.ceca.2023.102836.
[72] Bouyahya A, El Hachlafi N, et al. Natural Bioactive Compounds Targeting Histone Deacetylases in Human Cancers: Recent Updates, Molecules (2022) 27(8):2568. https://doi.org/10.3390/molecules27082568
[73] Zhixiang Wang, et al. Berberine acts as a putative epigenetic modulator by affecting the histone code, Toxicology in Vitro Volume 36 (2016) https://doi.org/10.1016/j.tiv.2016.06.004.
[74] Meyer, Stefanie N. et al. Unique and Shared Epigenetic Programs of the CREBBP and EP300 Acetyltransferases in Germinal Center B Cells Reveal Targetable Dependencies in Lymphoma, Immunity Volume 51, Issue 3 (2019) doi.org/10.1016/j.immuni.2019.08.006
[75] Lee, KH., Lo, HL., Tang, WC. et al. A gene expression signature-based approach reveals the mechanisms of action of the Chinese herbal medicine berberine, Sci Rep 4 (2014). https://doi.org/10.1038/srep06394
[76] Eustace AJ, Lee MJ, et al. Aberrant calcium signalling downstream of mutations in TP53 and the PI3K/AKT pathway genes promotes disease progression and therapy resistance in triple negative breast cancer. Cancer Drug Resist. (2022) http://dx.doi.org/10.20517/cdr.2022.41
[77] Shirakami Y, Shimizu M. Possible Mechanisms of Green Tea and Its Constituents against Cancer, Molecules Volume 23, Issue 9 https://doi.org/10.3390/molecules23092284
[78] Ciesielski O, Biesiekierska M, Balcerczyk A. Epigallocatechin-3-gallate (EGCG) Alters Histone Acetylation and Methylation and Impacts Chromatin Architecture Profile in Human Endothelial Cells, Molecules Volume 25, Issue 10 https://doi.org/10.3390/molecules25102326
[79] Gujral, P., Mahajan, V., Lissaman, A.C. et al. Histone acetylation and the role of histone deacetylases in normal cyclic endometrium, Reprod Biol Endocrinol Volume 18, Article 84 (2020) https://doi.org/10.1186/s12958-020-00637-5
[80] Yang, J., Nie, J., Ma, X. et al. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer 18, 26 (2019). https://doi.org/10.1186/s12943-019-0954-x
[81] Rachel L. Yamnik, et al. S6 Kinase 1 Regulates Estrogen Receptor α in Control of Breast Cancer Cell Proliferation*, Journal of Biological Chemistry, Volume 284, Issue 10, (2009) https://doi.org/10.1074/jbc.M807532200.
Ethical and Intellectual Stewardship
This work was born not in a lab but in a crisis — under the most profound personal loss. As such, the author, M. Malone, affirms that the underlying methodology is provisionally patented to ensure responsible development, not be co-opted for commercial exploitation detached from its ethical origins. The purpose of this discovery is to advance human health, not to convert tragedy into third-party profit. Any future use of this system must uphold rigorous ethical oversight, compassionate application, and the original intent: to save lives, improve quality of life, not monetize them.
Declarations
Author declares no conflict of interest
Acknowledgments
I wish to acknowledge the bravery and cooperation of the patient, wife & mother Jill, who had the courage to face the most difficult disease prognosis with innovation and adaptation when all hope seemed lost. Their contribution is the most integral to the research presented in this paper and will go on to further improve oncological research and genetic treatment options.
Funding
The study was self-funded by the patient and author M. Malone, and no third-parties were involved in this clinically approached patient case study.
Author contributions
M. Malone conceived, designed, administered and analyzed all aspects of this treatment protocol and its manuscript. Additional authors, Dr. Wimbs and Dr. Greer, provided clinical review and support of publication and were not involved in the patient’s treatment. This breakthrough also couldn’t have been achieved without the support and advocacy of the oncologists and specialists for prescriptions and monitoring results alongside in real-time. This accounts to several in-person, medical profession peer-reviews already on record prior to any journal publication.
Competing interests
Author declares no competing interests.
Ethics approval and Consent to participate
Treatment was voluntary by the patient, all prescriptions in the patient’s name, and not part of a research project, denoting N-of-1 compassionate use.
Consent to publish
Legal & ethical owner of the data is the author M. Malone, and has consented to publishing for furthering cancer research and genetic disease treatment innovation.
Data availability
Snippets of clinical reports are available to view, utilized throughout the paper, original reports are only available under exceptional circumstances to protect patient’s privacy.