1. What are the major hallmarks of cancer and how do they contribute to tumor progression?

Answer: The major hallmarks of cancer, as identified by researchers, include:

  • Sustaining proliferative signaling: Cancer cells can continuously signal themselves and surrounding cells to promote their own growth.
  • Evading growth suppressors: Tumor suppressor genes, such as p53 and Rb, are often mutated, allowing uncontrolled cell growth.
  • Resisting cell death: Cancer cells evade apoptosis, ensuring survival despite genetic abnormalities.
  • Enabling replicative immortality: Cancer cells acquire the ability to maintain telomere length, allowing them to divide indefinitely.
  • Inducing angiogenesis: Tumors stimulate the formation of new blood vessels, ensuring adequate oxygen and nutrient supply.
  • Activating invasion and metastasis: Cancer cells gain the ability to spread to distant organs.

These characteristics contribute to cancer’s ability to grow uncontrollably, evade the immune system, and spread throughout the body.


2. Describe the role of oncogenes and tumor suppressor genes in cancer development.

Answer:

  • Oncogenes are mutated versions of normal genes (proto-oncogenes) that promote cell growth and survival. When these genes are activated, they can drive tumor formation by stimulating abnormal cell division. Examples include the RAS gene, which encodes a protein involved in cell signaling pathways that control growth, and MYC, which regulates cell cycle progression.
  • Tumor suppressor genes act to prevent uncontrolled cell growth by either repairing damaged DNA or initiating cell death. Mutations in these genes can lead to loss of function, allowing abnormal cells to proliferate. A prominent example is p53, a protein that triggers apoptosis in response to DNA damage. RB is another key tumor suppressor gene involved in controlling cell cycle progression.

The balance between the activation of oncogenes and the inactivation of tumor suppressor genes is crucial for the development of cancer.


3. How do cancer cells evade the immune system?

Answer: Cancer cells can evade immune detection through several mechanisms:

  • Expression of immune checkpoint proteins: Proteins like PD-L1 are often expressed on the surface of cancer cells, which interact with the PD-1 receptor on T-cells, inhibiting the immune response.
  • Loss of tumor antigen presentation: Cancer cells can reduce the expression of major histocompatibility complex (MHC) molecules, making it difficult for immune cells to recognize tumor antigens.
  • Immunosuppressive microenvironment: Tumors can create an environment rich in immunosuppressive molecules like TGF-β and IL-10, which inhibit the activity of immune cells, such as T-cells and NK cells.

These mechanisms allow cancer cells to avoid destruction by the immune system, contributing to their ability to grow and spread.


4. What are the key advancements in targeted therapies for cancer treatment?

Answer: Targeted therapies are designed to specifically attack cancer cells while sparing normal cells. Key advancements include:

  • Tyrosine kinase inhibitors (TKIs): These drugs block the activity of tyrosine kinases, which are involved in cell signaling that drives tumor growth. For example, Imatinib is used to treat chronic myelogenous leukemia (CML) by inhibiting the BCR-ABL fusion protein.
  • Monoclonal antibodies: These antibodies target specific proteins on cancer cell surfaces. Trastuzumab (Herceptin) targets HER2, a receptor often overexpressed in breast cancer.
  • Small molecule inhibitors: Drugs like BRAF inhibitors are used to target specific mutated proteins, such as those found in melanoma with the BRAF V600E mutation.

These therapies have shown promising results, offering more precise treatment options and fewer side effects compared to traditional chemotherapy.


5. How does immunotherapy work in cancer treatment?

Answer: Immunotherapy uses the body’s immune system to fight cancer. There are several approaches:

  • Checkpoint inhibitors: Drugs like nivolumab and pembrolizumab block immune checkpoint proteins (PD-1 and PD-L1), which cancer cells use to evade the immune system.
  • CAR-T cell therapy: This involves modifying a patient’s T-cells to express chimeric antigen receptors (CARs) that recognize and target cancer cells. CAR-T therapy has shown remarkable success in hematologic cancers like leukemia and lymphoma.
  • Cancer vaccines: Vaccines such as the HPV vaccine (to prevent cervical cancer) and sipuleucel-T (used in prostate cancer) stimulate the immune system to recognize and destroy cancer cells.

Immunotherapy can enhance the immune system’s ability to recognize and kill cancer cells, providing long-lasting responses in some cases.


6. Explain the role of angiogenesis in cancer progression and how it is targeted in therapies.

Answer: Angiogenesis is the process of new blood vessel formation. Tumors require an adequate blood supply to grow and spread, so they secrete pro-angiogenic factors like vascular endothelial growth factor (VEGF). By stimulating angiogenesis, tumors can receive the nutrients and oxygen needed for continued growth.

  • Anti-angiogenic therapies aim to block the formation of blood vessels that supply the tumor. For example, Bevacizumab (Avastin) is a monoclonal antibody that targets VEGF and inhibits angiogenesis.

By blocking the tumor’s ability to form blood vessels, anti-angiogenesis therapies can starve the tumor, limiting its growth and spread.


7. What are the challenges associated with cancer stem cells in cancer therapy?

Answer: Cancer stem cells (CSCs) are a subset of cancer cells with the ability to self-renew and drive tumor growth. These cells are often more resistant to conventional therapies, including chemotherapy and radiation, due to:

  • Enhanced DNA repair mechanisms
  • Higher expression of drug efflux pumps
  • Quiescence, which makes them less susceptible to treatments targeting proliferating cells

Targeting CSCs has become a major area of research, as eliminating these cells could potentially prevent relapse and metastasis. However, effective therapies against CSCs are still under development.


8. What is the significance of genetic mutations in cancer and how are they detected?

Answer: Genetic mutations are at the core of cancer development, leading to changes in cell behavior. These mutations can occur in proto-oncogenes, tumor suppressor genes, or genes involved in DNA repair.

  • Next-generation sequencing (NGS) is a powerful tool for detecting mutations in cancer genomes, allowing researchers and clinicians to identify specific mutations that drive cancer.
  • Liquid biopsy is a non-invasive method that detects circulating tumor DNA (ctDNA) in blood samples, helping in the detection of mutations without the need for tissue biopsy.

By identifying these mutations, clinicians can tailor treatments to target specific alterations in the tumor’s DNA.


9. Describe the role of p53 in cancer biology and how its loss contributes to tumor progression.

Answer: The protein p53 is known as the “guardian of the genome” because it plays a critical role in maintaining genomic stability. It functions by:

  • Inducing cell cycle arrest to allow time for DNA repair.
  • Activating apoptosis if the damage is irreparable.
  • Inhibiting angiogenesis and metastasis.

Loss of p53 function, through mutations or deletions, is one of the most common alterations in cancer cells. Without functional p53, cells with damaged DNA can proliferate uncontrollably, leading to tumor formation and progression.


10. What is the role of the tumor microenvironment in cancer progression and how can it be targeted therapeutically?

Answer: The tumor microenvironment (TME) consists of cancer cells, stromal cells, immune cells, blood vessels, and extracellular matrix components. It plays a significant role in:

  • Promoting tumor growth through growth factors and cytokines.
  • Supporting angiogenesis and immune evasion.
  • Contributing to metastasis by remodeling the extracellular matrix.

Therapeutic strategies targeting the TME include:

  • Targeting immune suppressive cells like regulatory T-cells and myeloid-derived suppressor cells.
  • Inhibiting stromal cells that support tumor growth and metastasis.
  • Blocking the ECM remodeling to prevent tumor cell migration.

Targeting the TME can enhance the effectiveness of conventional cancer therapies and prevent relapse.


11. What is the significance of BRCA1 and BRCA2 mutations in cancer and how are they targeted in treatment?

Answer: Mutations in BRCA1 and BRCA2 genes are linked to an increased risk of breast, ovarian, and other cancers. These genes are involved in DNA repair through homologous recombination.

  • PARP inhibitors, such as Olaparib, are used to target cancer cells with BRCA mutations by inhibiting an alternative DNA repair pathway, leading to the accumulation of DNA damage and cell death.
  • Genetic testing for BRCA mutations helps in risk assessment and personalized treatment, including preventive measures and targeted therapies.

These advancements in targeting DNA repair mechanisms have improved outcomes for patients with BRCA-related cancers.


12. Discuss the role of epigenetics in cancer development and therapy.

Answer: Epigenetic changes refer to modifications in gene expression without altering the underlying DNA sequence. These changes include DNA methylation, histone modification, and non-coding RNA regulation. In cancer:

  • Hypermethylation of tumor suppressor genes can silence their expression, while hypomethylation can activate oncogenes.
  • Histone modifications can also alter chromatin structure, leading to the activation of oncogenes or silencing of tumor suppressors.

Epigenetic therapies aim to reverse these changes. For example, DNA demethylating agents and histone deacetylase inhibitors (HDAC inhibitors) are being explored in clinical trials to restore normal gene expression and prevent cancer progression.


13. What is the role of the Warburg effect in cancer metabolism?

Answer: The Warburg effect refers to the observation that cancer cells preferentially use glycolysis for energy production, even in the presence of oxygen, a process known as aerobic glycolysis. This is in contrast to normal cells, which rely on oxidative phosphorylation for energy production.

  • Cancer cells shift their metabolism to support rapid growth and survival in a hypoxic microenvironment.
  • The Warburg effect provides cancer cells with intermediates needed for biosynthesis and helps evade cell death.

Targeting cancer metabolism by inhibiting glycolysis or other metabolic pathways is an emerging therapeutic strategy.


14. What is the role of metastasis in cancer and how can it be prevented?

Answer: Metastasis is the process by which cancer cells spread from the primary tumor to distant organs, making cancer more difficult to treat. The steps involved in metastasis include:

  • Invasion of surrounding tissue.
  • Intravasation into blood vessels or lymphatics.
  • Survival in circulation.
  • Extravasation into distant tissues.
  • Colonization and growth in new tissues.

Preventing metastasis involves targeting the molecular mechanisms responsible for these processes. Therapies that inhibit metastasis are still in development but show promise in disrupting the ability of cancer cells to spread.


15. What is the concept of personalized medicine in cancer treatment and how does it improve patient outcomes?

Answer: Personalized medicine involves tailoring treatment based on the individual genetic profile of the patient’s tumor. This approach considers the specific mutations and characteristics of the cancer, allowing for targeted therapies that are more effective and less toxic.

  • Genomic profiling and liquid biopsy help identify actionable mutations.
  • Targeted therapies are then selected to match the specific mutation, such as EGFR inhibitors for non-small cell lung cancer.

Personalized medicine improves patient outcomes by increasing the likelihood of treatment success and minimizing side effects.


16. Explain the role of tumor microRNA in cancer and its potential for therapeutic targeting.

Answer: MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression by binding to messenger RNAs. In cancer:

  • OncomiRs are miRNAs that promote tumorigenesis by inhibiting tumor suppressor genes.
  • Tumor-suppressive miRNAs can be downregulated in cancer, allowing uncontrolled growth.

Targeting miRNAs as a therapeutic approach involves restoring the function of tumor-suppressive miRNAs or inhibiting oncomiRs. This area of research is still in early stages but holds potential for cancer therapy.


17. What is the significance of liquid biopsy in cancer diagnosis and monitoring?

Answer: Liquid biopsy is a non-invasive technique used to detect genetic material from tumor cells in bodily fluids such as blood or urine. It is used for:

  • Early detection of cancer by identifying tumor markers or ctDNA.
  • Monitoring treatment response and detecting minimal residual disease or recurrence.
  • Identifying genetic mutations that could guide personalized therapy.

Liquid biopsy offers advantages over traditional tissue biopsy, such as being less invasive and allowing real-time monitoring of the tumor’s genetic landscape.


18. How do DNA repair inhibitors work in cancer therapy?

Answer: DNA repair inhibitors target the ability of cancer cells to repair DNA damage, thereby enhancing the effectiveness of chemotherapy and radiation. Examples include:

  • PARP inhibitors: These block the repair of single-strand breaks and are especially effective in cancers with defective homologous recombination (e.g., BRCA-mutated cancers).
  • ATR inhibitors: These target the ATR pathway, which responds to DNA damage during the cell cycle.

By inhibiting DNA repair, these therapies cause cancer cells to accumulate lethal DNA damage, improving treatment outcomes.


19. What are the implications of drug resistance in cancer therapy and how can it be overcome?

Answer: Drug resistance occurs when cancer cells adapt to survive despite the presence of therapeutic agents. Mechanisms include:

  • Mutations that alter the drug target.
  • Increased drug efflux or reduced drug uptake.
  • Activation of alternative signaling pathways. To overcome drug resistance, strategies include:
  • Combination therapies that target multiple pathways simultaneously.
  • Targeting drug resistance mechanisms using inhibitors or immunotherapies.
  • Developing next-generation drugs that can overcome specific mutations.

Ongoing research aims to understand resistance mechanisms better and develop drugs that can effectively address these challenges.


20. How has the development of CRISPR-Cas9 revolutionized cancer research and treatment?

Answer: The CRISPR-Cas9 gene-editing technology allows precise modifications of DNA, enabling researchers to:

  • Knock out genes to study their role in cancer development.
  • Correct genetic mutations associated with cancer.
  • Enhance the immune response by editing T-cells in immunotherapy.

CRISPR has the potential to not only advance our understanding of cancer biology but also create personalized treatments by editing specific mutations or enhancing the immune system’s ability to target tumors.

LEAVE A REPLY

Please enter your comment!
Please enter your name here