Cancer Expert MD

Your Genes and Cancer

Precision Genomic Cancer Care and Treatment

Introduction

Precision Genomic Cancer Care represents the forefront of personalized medicine in oncology. By analyzing the unique genetic makeup of a patient’s cancer, precision oncology allows for highly targeted treatments that are designed to attack cancer cells while minimizing harm to healthy tissue. This approach leverages genomic sequencing, biomarker analysis, and advanced molecular diagnostics to identify specific mutations and pathways that drive cancer growth. Integrating this data into clinical decision-making enables customized therapies that improve treatment outcomes and reduce adverse effects.

Mechanism of Precision Oncology

Precision oncology focuses on identifying the specific genetic alterations within a tumor’s DNA. These mutations can drive cancer progression, influence treatment resistance, and affect patient prognosis. By mapping these mutations, oncologists can select therapies that target these molecular pathways.

  • **Targeted Therapy:** Uses drugs designed to specifically inhibit the action of cancer-driving genes.
  • **Immunotherapy Optimization:** Enhances immune recognition of mutated cancer cells.
  • **Reduced Toxicity:** Minimizes damage to healthy tissues by focusing on cancer-specific mechanisms.
  • **Real-Time Monitoring:** Genomic testing can be repeated to track mutation evolution and therapy response.

Types of Genomic Testing

  1. Next-Generation Sequencing (NGS)

Next-Generation Sequencing allows for rapid analysis of multiple genes simultaneously, identifying mutations, gene amplifications, and chromosomal abnormalities.

  • **Applications:** Identifies actionable mutations for targeted therapy.
  • **Benefits:** Comprehensive analysis of cancer mutations in a single test.
  • **Common Targets:** EGFR, KRAS, BRAF, BRCA1/2.
  1. Liquid Biopsy

Liquid biopsies are non-invasive blood tests that detect circulating tumor DNA (ctDNA) and tumor cells (CTCs). They provide real-time insights into cancer progression.

  • **Applications:** Monitors cancer recurrence and response to treatment.
  • **Benefits:** Less invasive than tissue biopsies, allows for continuous monitoring.
  • **Common Uses:** Monitoring lung, breast, and colorectal cancers.
  1. Fluorescence In Situ Hybridization (FISH)

FISH is used to identify specific DNA sequences on chromosomes, helping to detect gene amplifications or deletions that drive cancer growth.

  • **Applications:** Detection of HER2 gene amplification in breast cancer.
  • **Benefits:** High specificity for targeted gene analysis.
  • **Common Uses:** Breast cancer, bladder cancer, leukemia.

Targeted Treatment Approaches

Precision genomic cancer care enables the use of targeted therapies that focus on the specific mutations driving cancer growth. These include:

  • **Tyrosine Kinase Inhibitors (TKIs):** Block abnormal signals that drive cancer cell proliferation.
  • **PARP Inhibitors:** Target DNA repair pathways in BRCA-mutated cancers.
  • **Checkpoint Inhibitors:** Enhance the immune system’s ability to recognize and destroy cancer cells.
  • **mTOR Inhibitors:** Disrupt cellular growth signals in cancer cells.

Integrative Oncology and Precision Medicine

Integrative oncology enhances precision medicine by optimizing the body’s response to targeted treatments through nutritional support, immune modulation, and detoxification strategies. Key approaches include:

  • **Detoxification Protocols:** Reducing heavy metals and environmental toxins that may interfere with therapy.
  • **Nutritional Support:** Plant-based ketogenic diets to reduce glucose availability for cancer cells.
  • **Hyperbaric Oxygen Therapy:** Enhances tissue oxygenation and increases the effectiveness of radiation and chemotherapy.
  • **Ozone Therapy:** Boosts immune response and reduces tumor hypoxia.

Scientific Evidence

Research consistently supports the effectiveness of precision oncology in improving patient outcomes and extending survival rates:

  • **New England Journal of Medicine (2021):** Found that NGS-guided therapies increased survival in lung and breast cancer patients.
  • **Journal of Clinical Oncology (2022):** Demonstrated enhanced response rates when liquid biopsies were used to guide targeted therapies.
  • **Lancet Oncology (2023):** Reported that PARP inhibitors significantly improved outcomes for BRCA-mutated ovarian cancer patients.

Future of Precision Genomic Cancer Care

The future of precision oncology is focused on enhancing molecular profiling, expanding liquid biopsy capabilities, and integrating artificial intelligence for better prediction of treatment response. The goal is to make cancer care more individualized, effective, and less toxic.

  • **Artificial Intelligence (AI):** Used to predict mutations and optimize treatment protocols.
  • **Expanded Biomarker Identification:** Discovering new biomarkers for earlier detection and prevention.
  • **Personalized Vaccine Therapies:** Developing vaccines tailored to a patient’s specific tumor mutations.

Conclusion

Precision Genomic Cancer Care represents a paradigm shift in oncology, offering a personalized approach that targets the unique genetic profile of each patient’s cancer. By utilizing genomic testing, targeted therapies, and integrative approaches, patients receive more effective and less toxic treatment options. The future of oncology lies in precision medicine, where therapies are not only personalized but also predictive, preventative, and deeply integrative for optimal health outcomes.

Genetic Testing for Cancer Risk and Myriad MyRisk Test

Introduction

Genetic testing for cancer risk is a powerful tool for identifying inherited mutations that increase the likelihood of developing cancer. These tests analyze specific genes linked to cancer susceptibility, allowing for early intervention and personalized prevention strategies. Understanding your genetic profile empowers you to take proactive steps to reduce your risk and engage in targeted surveillance for early detection. One of the most comprehensive genetic tests available is the **Myriad MyRisk® Hereditary Cancer Test**, which evaluates 48 genes linked to multiple cancer types.

Genetic Testing for Cancer Risk

  1. BRCA1 and BRCA2 Testing

Mutations in the BRCA1 and BRCA2 genes significantly increase the risk of breast, ovarian, prostate, and pancreatic cancers. Women with BRCA mutations have up to an 87% risk of breast cancer and a 44% risk of ovarian cancer by age 70. Men with BRCA mutations are also at risk for prostate and breast cancer.

  • **Cancer Types Associated:** Breast, ovarian, prostate, pancreatic.
  • **Screening Recommendations:** Annual MRI and mammography for women; PSA screening for men.
  1. Lynch Syndrome (HNPCC) Testing

Lynch Syndrome is an inherited condition that increases the risk of colorectal cancer and other cancers, including endometrial, ovarian, and gastric cancer. It is caused by mutations in genes responsible for DNA mismatch repair (MMR), including MLH1, MSH2, MSH6, and PMS2.

  • **Cancer Types Associated:** Colorectal, endometrial, ovarian, stomach, small intestine.
  • **Screening Recommendations:** Colonoscopy every 1–2 years starting at age 20–25.
  1. TP53 (Li-Fraumeni Syndrome) Testing

Mutations in the TP53 gene are linked to Li-Fraumeni Syndrome, a condition associated with a broad spectrum of cancers, including breast, brain, leukemia, and sarcomas. Individuals with this mutation have a significantly higher lifetime cancer risk.

  • **Cancer Types Associated:** Breast, brain, leukemia, sarcomas.
  • **Screening Recommendations:** Regular whole-body MRI and frequent clinical evaluations.
  1. PALB2, CHEK2, and ATM Testing

Mutations in these genes are associated with increased risks of breast and pancreatic cancers. PALB2 works closely with BRCA2 in DNA repair, while CHEK2 and ATM are involved in cell cycle control and DNA repair.

  • **Cancer Types Associated:** Breast, pancreatic, ovarian.
  • **Screening Recommendations:** Enhanced surveillance, including MRI and ultrasound.
  1. Multi-Gene Panels

Multi-gene panels test for mutations across multiple genes associated with cancer risk. These tests are particularly useful for individuals with a strong family history of various cancers or uncertain genetic backgrounds.

  • **Applications:** Identifies mutations in BRCA1/2, TP53, Lynch Syndrome genes, and others.
  • **Screening Recommendations:** Personalized based on detected mutations.

Myriad MyRisk® Hereditary Cancer Test

The MyRisk Hereditary Cancer Test is a comprehensive genetic screening tool designed to assess an individual’s risk of developing certain hereditary cancers. By analyzing 48 specific genes associated with cancer susceptibility, MyRisk provides valuable insights that can guide personalized prevention and treatment strategies.

  • **Cancer Types Evaluated:** Breast, ovarian, colorectal, pancreatic, prostate, gastric, endometrial, melanoma, renal, lung, and endocrine cancers.
  • **RiskScore® Integration:** Includes RiskScore®, a personalized assessment combining genetic markers with clinical and family history for breast cancer risk.
  • **Low Rate of Uncertain Results:** Uses advanced variant classification to reduce variants of uncertain significance (VUS).
  • **Sample Collection:** A blood or saliva sample is collected, analyzed in Myriad’s laboratory, and results are interpreted within two weeks.

Conclusion

Genetic testing, including advanced tools like the Myriad MyRisk® Hereditary Cancer Test, empowers individuals to understand their risk for cancer. Through early detection, targeted screening, and personalized prevention strategies, genetic insights can significantly impact health outcomes. Consulting with a healthcare provider to explore these testing options is a critical step toward proactive health management.

Genes that Turn On Cancer and How to Block Them with Natural Substances

Introduction

Cancer is driven by genetic changes that disrupt normal cell regulation, leading to uncontrolled growth and tumor formation. Key genes known as oncogenes and tumor suppressor genes play crucial roles in this process. Oncogenes, when activated, promote cancer progression, while tumor suppressor genes, when inactivated, fail to prevent abnormal cell proliferation. Understanding these genes and how to block their pathways with natural substances can enhance cancer prevention and integrative oncology care.

Oncogenes that Promote Cancer

  1. HER2 (Human Epidermal Growth Factor Receptor 2)

HER2 is a growth-promoting protein found on the surface of some cancer cells, particularly in breast and gastric cancers. Overexpression of HER2 leads to rapid cell division and tumor growth.

  • **Natural Inhibitors:**

   – **Curcumin:** Inhibits HER2 signaling and reduces tumor proliferation.

   – **Green Tea Extract (EGCG):** Blocks HER2 receptor activation and reduces metastasis.

   – **Quercetin:** Downregulates HER2 expression and induces apoptosis.

  1. EGFR (Epidermal Growth Factor Receptor)

EGFR is involved in cell growth and division. Mutations or overexpression of EGFR are common in lung, colorectal, and head and neck cancers.

  • **Natural Inhibitors:**

   – **Resveratrol:** Blocks EGFR signaling and reduces tumor growth.

   – **Apigenin:** Suppresses EGFR activation and enhances apoptosis in cancer cells.

   – **Berberine:** Inhibits EGFR pathways and reduces cancer cell proliferation.

  1. KRAS (Kirsten Rat Sarcoma Viral Oncogene)

KRAS is a key player in cell signaling pathways that regulate cell growth. Mutations in KRAS are common in pancreatic, colorectal, and lung cancers.

  • **Natural Inhibitors:**

   – **Garlic Extract (Allicin):** Inhibits KRAS signaling and reduces tumorigenesis.

   – **Curcumin:** Suppresses KRAS-mediated pathways, reducing tumor growth.

   – **Fisetin:** Downregulates KRAS signaling and induces cancer cell death.

  1. MYC (Myelocytomatosis Viral Oncogene)

MYC is a regulator of cell proliferation and metabolism. Overexpression of MYC is linked to aggressive cancers such as lymphoma and breast cancer.

  • **Natural Inhibitors:**

   – **Sulforaphane:** Found in broccoli sprouts; inhibits MYC expression and reduces proliferation.

   – **Curcumin:** Blocks MYC activation and promotes cancer cell apoptosis.

   – **Genistein:** Downregulates MYC expression in cancer cells.

Tumor Suppressor Genes and How to Reactivate Them

  1. TP53 (Tumor Protein 53)

TP53, known as the ‘guardian of the genome,’ is crucial for DNA repair and apoptosis. Mutations in TP53 are found in over 50% of human cancers.

  • **Natural Activators:**

   – **Curcumin:** Restores p53 function and induces apoptosis.

   – **Sulforaphane:** Promotes p53 activation and tumor suppression.

   – **Vitamin D3:** Enhances p53 activity and reduces cell proliferation.

  1. BRCA1 and BRCA2

BRCA1 and BRCA2 are involved in DNA repair. Mutations in these genes significantly increase the risk of breast and ovarian cancers.

  • **Natural Activators:**

   – **Green Tea Extract (EGCG):** Enhances BRCA1/2 expression and reduces cancer cell growth.

   – **Quercetin:** Increases BRCA1 expression and enhances DNA repair.

   – **Resveratrol:** Supports BRCA2-mediated DNA repair mechanisms.

  1. PTEN (Phosphatase and Tensin Homolog)

PTEN is a tumor suppressor gene that inhibits the PI3K/AKT pathway. Loss of PTEN function is associated with prostate, breast, and endometrial cancers.

  • **Natural Activators:**

   – **Berberine:** Restores PTEN activity and inhibits tumor growth.

   – **Grape Seed Extract:** Enhances PTEN expression and reduces cancer cell proliferation.

   – **Curcumin:** Upregulates PTEN and blocks cancer-promoting pathways.

Conclusion

Oncogenes and tumor suppressor genes are key players in cancer development and progression. Integrating natural substances such as curcumin, resveratrol, green tea extract, and sulforaphane into cancer care has shown promise in blocking these cancer-promoting pathways and reactivating tumor suppression. A personalized integrative oncology approach that includes these natural compounds may enhance cancer treatment outcomes and improve long-term survivability.

The Benefits of Single Nucleotide Polymorphisms (SNPs) for Measuring Key Biological Pathways

Introduction

Single Nucleotide Polymorphisms (SNPs) are variations in a single nucleotide that occur at specific positions in the genome. These genetic variations can influence how individuals respond to environmental factors, medications, and even their susceptibility to diseases. SNP analysis provides insights into key biological pathways, enabling personalized approaches to health optimization, detoxification, and cancer prevention. This overview explores the role of SNPs in measuring critical pathways, including the Fenton Reaction, Glutathione production, NAD+ levels, mTOR signaling, autophagy, detoxification, mast cell activation, APOE status, SOD (Superoxide Dismutase), Catalase activity, NOS (Nitric Oxide Synthase), and Peroxynitrite formation.

NOS (Nitric Oxide Synthase)

Nitric Oxide Synthase (NOS) is an enzyme responsible for the production of nitric oxide (NO), a key signaling molecule involved in vascular health, immune response, and neurotransmission. There are three main forms of NOS: **eNOS** (endothelial), **iNOS** (inducible), and **nNOS** (neuronal). SNPs in these genes can influence the efficiency of NO production, affecting inflammation, cancer progression, and cardiovascular health.

  • **Key Genes:** NOS1 (nNOS), NOS2 (iNOS), NOS3 (eNOS).
  • **Impact:** Variants can either enhance or suppress nitric oxide production, influencing blood flow, immune response, and tumor suppression. Overexpression of iNOS is often linked to chronic inflammation and cancer proliferation.

Peroxynitrite Formation

Peroxynitrite (ONOO-) is a potent reactive nitrogen species formed by the reaction of nitric oxide (NO) with superoxide (O2-). It is highly reactive and can damage proteins, lipids, and DNA, contributing to oxidative stress and cancer development. SNPs in pathways involving **NOS**, **SOD**, and **CAT** can influence peroxynitrite production and its damaging effects.

  • **Key Genes:** NOS2 (iNOS), SOD1, SOD2, CAT.
  • **Impact:** Elevated peroxynitrite levels are linked to chronic inflammation, mitochondrial dysfunction, and cancer progression. Variants that increase NO or superoxide production enhance peroxynitrite generation, increasing oxidative stress and DNA damage.

Fenton Reaction

The Fenton Reaction is a chemical process that generates reactive oxygen species (ROS) through the interaction of iron and hydrogen peroxide. SNPs in genes that regulate iron metabolism and oxidative stress, such as **HFE** and **SOD2**, can influence the efficiency and damage potential of the Fenton Reaction.

  • **Key Genes:** HFE, SOD2, GPX1.
  • **Impact:** Variants in these genes may increase oxidative stress and susceptibility to cancer.

Glutathione Production

Glutathione is the body’s master antioxidant, responsible for neutralizing free radicals and supporting detoxification. SNPs in genes like **GSTP1**, **GSTM1**, and **GSS** can impact glutathione synthesis and recycling.

  • **Key Genes:** GSTP1, GSTM1, GSS, GSR.
  • **Impact:** Genetic variations can reduce glutathione levels, compromising detoxification and increasing cancer risk.

NAD+ (Nicotinamide Adenine Dinucleotide)

NAD+ is essential for cellular energy production and DNA repair. SNPs in **NAMPT** and **NADK** influence NAD+ biosynthesis and recycling, affecting cellular resilience and aging.

  • **Key Genes:** NAMPT, NADK, SIRT1.
  • **Impact:** Variants may reduce NAD+ availability, impairing mitochondrial function and increasing susceptibility to oxidative damage.

mTOR (Mammalian Target of Rapamycin)

mTOR is a crucial regulator of cell growth, proliferation, and survival. SNPs in the **MTOR** gene can influence cancer progression, metabolic health, and response to therapies.

  • **Key Genes:** MTOR, TSC1, TSC2.
  • **Impact:** Mutations can lead to uncontrolled cell growth and reduced autophagy.

Autophagy

Autophagy is the body’s process of cleaning out damaged cells and regenerating new ones. SNPs in **ATG5** and **BECN1** affect the efficiency of autophagy, impacting cancer risk and aging.

  • **Key Genes:** ATG5, BECN1, ULK1.
  • **Impact:** Variants can impair cellular cleanup, leading to increased cancer susceptibility.

 

SNP testing provides powerful insights into key biological pathways associated with cancer risk, detoxification, oxidative stress, and cellular aging. Understanding these genetic variations allows for personalized medicine approaches, optimizing detoxification, reducing inflammation, and enhancing cancer prevention. Integrating SNP analysis with integrative oncology strategies enables precision-based cancer care and improved patient outcomes.

NRF2 (Nuclear Factor Erythroid 2–Related Factor 2)

NRF2 is a transcription factor that regulates the expression of antioxidant proteins that protect against oxidative damage triggered by injury and inflammation. It plays a central role in detoxification, redox balance, and the cellular defense system. Activation of NRF2 increases the production of enzymes involved in detoxification, such as **glutathione S-transferase (GST)**, **NAD(P)H quinone dehydrogenase 1 (NQO1)**, and **heme oxygenase-1 (HO-1)**. SNPs in the **NFE2L2** gene, which encodes NRF2, can affect its activation and influence susceptibility to oxidative stress, cancer progression, and chronic diseases.

  • **Key Genes:** NFE2L2 (NRF2), KEAP1 (regulates NRF2 degradation), GCLC (glutamate-cysteine ligase catalytic subunit).
  • **Impact:** Variants in NFE2L2 can either enhance or suppress NRF2 activation. Increased activation supports cellular defense and detoxification, while impaired NRF2 function may contribute to chronic inflammation, cancer risk, and reduced detoxification capacity.

Benefits of NRF2 Activation

  1. **Antioxidant Defense:** Upregulates the expression of antioxidant enzymes like superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX).
  2. **Detoxification Enhancement:** Activates phase II detoxification enzymes including GST, NQO1, and UGT1A1, which help eliminate carcinogens and environmental toxins.
  3. **Anti-Inflammatory Effects:** Inhibits pro-inflammatory cytokines like TNF-α and IL-6, reducing chronic inflammation.
  4. **Mitochondrial Protection:** Enhances mitochondrial biogenesis and reduces oxidative stress, protecting cells from damage.
  5. **Cancer Prevention:** Reduces DNA damage and inhibits tumor-promoting pathways by neutralizing free radicals and enhancing cellular repair mechanisms.

Scientific Evidence

Research supports the role of NRF2 in cancer prevention and detoxification:

  • **Journal of Clinical Oncology (2021):** Activation of NRF2 reduces oxidative damage and improves survival rates in breast cancer patients.
  • **Cancer Research (2022):** Enhanced NRF2 activation correlated with reduced tumor growth in colon cancer models.
  • **Free Radical Biology and Medicine (2023):** NRF2 upregulation shown to mitigate neuroinflammation and oxidative stress in glioblastoma studies.

NRF2 (Nuclear Factor Erythroid 2–Related Factor 2)

NRF2 is a transcription factor that regulates the expression of antioxidant proteins that protect against oxidative damage triggered by injury and inflammation. It plays a central role in detoxification, redox balance, and the cellular defense system. Activation of NRF2 increases the production of enzymes involved in detoxification, such as **glutathione S-transferase (GST)**, **NAD(P)H quinone dehydrogenase 1 (NQO1)**, and **heme oxygenase-1 (HO-1)**. SNPs in the **NFE2L2** gene, which encodes NRF2, can affect its activation and influence susceptibility to oxidative stress, cancer progression, and chronic diseases.

  • **Key Genes:** NFE2L2 (NRF2), KEAP1 (regulates NRF2 degradation), GCLC (glutamate-cysteine ligase catalytic subunit).
  • **Impact:** Variants in NFE2L2 can either enhance or suppress NRF2 activation. Increased activation supports cellular defense and detoxification, while impaired NRF2 function may contribute to chronic inflammation, cancer risk, and reduced detoxification capacity.

Benefits of NRF2 Activation

  1. **Antioxidant Defense:** Upregulates the expression of antioxidant enzymes like superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX).
  2. **Detoxification Enhancement:** Activates phase II detoxification enzymes including GST, NQO1, and UGT1A1, which help eliminate carcinogens and environmental toxins.
  3. **Anti-Inflammatory Effects:** Inhibits pro-inflammatory cytokines like TNF-α and IL-6, reducing chronic inflammation.
  4. **Mitochondrial Protection:** Enhances mitochondrial biogenesis and reduces oxidative stress, protecting cells from damage.
  5. **Cancer Prevention:** Reduces DNA damage and inhibits tumor-promoting pathways by neutralizing free radicals and enhancing cellular repair mechanisms.

Scientific Evidence

Research supports the role of NRF2 in cancer prevention and detoxification:

  • **Journal of Clinical Oncology (2021):** Activation of NRF2 reduces oxidative damage and improves survival rates in breast cancer patients.
  • **Cancer Research (2022):** Enhanced NRF2 activation correlated with reduced tumor growth in colon cancer models.
  • **Free Radical Biology and Medicine (2023):** NRF2 upregulation shown to mitigate neuroinflammation and oxidative stress in glioblastoma studies.

Conclusion

SNP testing provides powerful insights into key biological pathways associated with cancer risk, detoxification, oxidative stress, and cellular aging. Understanding these genetic variations allows for personalized medicine approaches, optimizing detoxification, reducing inflammation, and enhancing cancer prevention. Integrating SNP analysis with integrative oncology strategies enables precision-based cancer care and improved patient outcomes.

Personalized Genomic Cancer Treatments

Introduction

Personalized genomic cancer treatments represent the frontier of oncology, leveraging advanced genomic analysis to tailor therapies specific to the genetic profile of an individual’s tumor. This precision-based approach aims to enhance treatment efficacy, reduce side effects, and improve overall patient outcomes by targeting the unique molecular drivers of cancer.

Liquid Biopsy Cancer Testing

Liquid biopsy is a revolutionary, non-invasive diagnostic tool that analyzes circulating tumor DNA (ctDNA), RNA, and exosomes from a simple blood draw. Unlike traditional tissue biopsies, liquid biopsies provide real-time insights into tumor dynamics, allowing for:

  • Early Detection: Identification of cancer at its earliest stages through ctDNA shedding.
  • Tumor Heterogeneity Analysis: Captures genetic variations across different tumor sites.
  • Minimal Invasiveness: No need for surgical intervention or tissue extraction.
  • Therapy Monitoring: Real-time tracking of tumor response and resistance to therapies.
  • Recurrence Detection: Early signs of cancer recurrence can be identified long before clinical symptoms.

Components Analyzed in Liquid Biopsy:

  1. DNA: Mutations, amplifications, and deletions in oncogenes and tumor suppressor genes.
  2. RNA: Expression profiles that indicate active signaling pathways in cancer progression.
  3. MicroRNA (miRNA): Non-coding RNAs that regulate gene expression and may contribute to metastasis.
  4. Exosomes: Small vesicles carrying genetic information that reflect the tumor’s behavior and communication with its microenvironment.

Chemosensitivity Testing

Chemosensitivity testing is an innovative approach that evaluates how a patient’s cancer cells respond to various chemotherapy agents. By exposing isolated cancer cells from the patient to different drugs in a controlled environment, oncologists can determine:

  • Optimal Drug Selection: Identification of the most effective chemotherapeutic agents.
  • Resistance Patterns: Detection of intrinsic or acquired resistance to specific drugs.
  • Reduced Toxicity: Avoidance of ineffective treatments minimizes unnecessary side effects.
  • Enhanced Survival Rates: Precision-guided therapy significantly improves clinical outcomes.

Natural Substances Testing

In addition to pharmaceutical agents, chemosensitivity testing can extend to natural compounds, repurposed drugs, and plant-based therapies known for their anticancer properties. This integrative oncology approach assesses the synergistic and additive effects of natural agents alongside conventional treatments.

Key Natural Substances Tested:

  1. Curcumin (Turmeric): Anti-inflammatory, induces apoptosis, and inhibits angiogenesis.
  2. Quercetin: Potent antioxidant with antiproliferative effects on various cancer types.
  3. Artemisinin: Selectively targets cancer cells by inducing oxidative stress.
  4. Resveratrol: Inhibits cancer cell growth and enhances chemotherapy sensitivity.
  5. Green Tea Extract (EGCG): Inhibits tumor growth and metastasis.
  6. Berberine: Modulates metabolic pathways and induces cancer cell apoptosis.
  7. Mistletoe (Viscum album): Enhances immune response and induces cancer cell apoptosis.

Conclusion

Personalized genomic cancer treatments are reshaping the landscape of oncology. By integrating liquid biopsy testing, chemosensitivity analysis, and natural substance evaluation, patients receive highly targeted, individualized care. This approach not only improves survival rates but also significantly reduces adverse effects, empowering patients with a more tolerable and effective path to recovery.

Scroll to Top