kembara Xtra
  • Introduction
  • Earth
  • Gemstones
  • Medicine
  • Finance
  • Law
  • Psychology
  • Technology
  • Travel The World
  • Who We Are
  • Get In Touch
  • Introduction
  • Earth
  • Gemstones
  • Medicine
  • Finance
  • Law
  • Psychology
  • Technology
  • Travel The World
  • Who We Are
  • Get In Touch

MEDICINE 

Oncology-Chemotherapeutic Dose Intensification in Oncology

4/29/2025

0 Comments

 
Oncology- Chemotherapeutic Dose Intensification in Oncology
This guide summarizes the provided text on dose intensification in cancer treatment, focusing on key concepts and clinical applications.
I. Dose-Response and Dose Intensification:
  • Central Idea: Cancer cell drug resistance is often relative, not absolute. Therefore, arbitrarily reducing drug doses should be avoided.
  • Clinical Implications: To overcome resistance and achieve better outcomes, dose intensification strategies are employed. This might necessitate using supportive care (e.g., prophylactic antibiotics, hematopoietic growth factors) to mitigate the increased toxicity associated with higher doses. The goal is to deliver potentially curative chemotherapy on a timely basis, even if it means managing side effects.
II. High-Dose Chemotherapy (HDC) with Hematopoietic Support:
  • Limitations of Conventional Dose Escalation: Increasing chemotherapy doses within a conventional range has inconsistently improved response rates and minimally impacted survival, often with increased toxicity.
  • Hematopoietic Support: Advances in hematopoietic support (using autologous bone marrow or cytokine-mobilized peripheral blood progenitors – PBPs) have enabled the use of HDC. These methods rescue the bone marrow from the myelosuppressive effects of the high-dose chemotherapy.
  • PBP Autografting: Superior to marrow autografting, leading to shorter periods of neutropenia and thrombocytopenia, reduced mortality, and reduced morbidity. This involves harvesting PBPs from the peripheral blood via leucopheresis, then re-infusing them after HDC.
  • Timing and Administration of HDC: Primarily used as consolidation therapy after conventional chemotherapy. Less commonly used as first-line (initial) treatment. Can be given in single or multiple cycles.
III. Role of HDC in Specific Cancers:
HDC has shown efficacy in various cancers, although the success rates vary:
  • Relapsed aggressive lymphoma: Proven salvage therapy (treatment after other therapies have failed).
  • Refractory lymphoma: Lower remission rates (around 10%).
  • Poor prognosis Non-Hodgkin lymphoma (NHL): May be used as a first-line treatment.
  • Multiple myeloma: May be used as a first-line treatment.
  • Relapsed/refractory Hodgkin's disease: May be used as a first-line treatment.
  • Acute leukemia: Especially relevant when a bone marrow donor is unavailable.
  • Metastatic testicular germ cell tumors: In cases of relapse after the second remission.
IV. Accelerated Chemotherapy:
  • Alternative to HDC: This strategy shortens the interval between cycles of conventional chemotherapy, typically supported by granulocyte colony-stimulating factor (G-CSF).
  • Status: Shows promise (particularly in adjuvant therapy for high-risk breast cancer), but remains experimental.
Key Terms to Understand:
  • Myelosuppression: Suppression of bone marrow function, leading to reduced blood cell production (neutropenia, thrombocytopenia).
  • Autografting: Transplantation of the patient's own bone marrow or PBPs.
  • Leucopheresis: Procedure to separate white blood cells (including PBPs) from blood.
  • Neutropenia: Low neutrophil count (type of white blood cell).
  • Thrombocytopenia: Low platelet count.
  • Adjuvant chemotherapy: Chemotherapy given after the primary treatment (surgery, radiation) to reduce the risk of recurrence.
  • Consolidation chemotherapy: Intensive chemotherapy given after achieving remission to further reduce the risk of recurrence.
  • Salvage therapy: Treatment given after initial treatment has failed.
  • Refractory: Resistant to treatment.
This study guide provides a structured overview of dose intensification strategies in oncology. Remember to review the original text for more detailed information and specific nuances. Focus on understanding the rationale behind dose intensification, the role of supportive care, and the specific applications of HDC and accelerated chemotherapy in various cancer types.


Picture
0 Comments

Oncology- Drug Resistance in Oncology

4/29/2025

0 Comments

 
Oncology- Drug Resistance in Oncology
This study guide summarizes the provided text on drug resistance in cancer treatment. Understanding these mechanisms is crucial for developing effective therapies.
I. Introduction: The Problem of Drug Resistance
  • Laboratory vs. Clinical Resistance: Much research uses artificially induced resistance in cell lines (often >40-100 fold increase in drug concentration needed to overcome resistance). The relevance of this to clinical resistance remains unclear.
II. Mechanisms of Drug Resistance
This section details the various ways cancer cells evade chemotherapy. Note that multiple mechanisms can operate simultaneously in a single patient.
A. Pharmacological Resistance: The effective drug concentration at the target site is insufficient due to:
  • Organ Toxicity: Limiting dosage due to side effects in other organs.
  • Enhanced Drug Clearance: The body removes the drug too quickly.
  • Physical Barriers: Tumour avascularity (lack of blood vessels) prevents drug delivery.
  • De Novo Resistance: The tumour is unresponsive to chemotherapy from the start.
  • Acquired Resistance: Initial response followed by tumour regrowth and resistance.
  • Combination of De Novo and Acquired: Both mechanisms are at play.
B. Alteration of Target or Transport Mechanisms: Tumour cells adapt by:
  • Reduced Drug Uptake: Mutations prevent drug entry into the cell.
  • Increased Detoxification: Faster metabolism and elimination of the drug.
  • Target Site Mutation: The drug's target is altered, making it ineffective.
  • Enhanced DNA Repair: Increased efficiency in repairing drug-induced damage.
C. Classical Multidrug Resistance (MDR):
  • P-170 Glycoprotein (Pgp): Overexpression of this 170kDa glycoprotein creates an energy-dependent drug efflux pump. Drugs enter the cell but are actively pumped back out, reducing intracellular concentration. This is common with anthracyclines, taxanes, and etoposide; resistance to one often implies resistance to others (multidrug resistance).
D. Multidrug Resistance-Associated Protein (MRP):
  • 190kDa Efflux Pump: Another energy-dependent pump causing drug efflux or sequestration within organelles. Substrate specificity similar to Pgp but generally associated with less taxane resistance. Clinical significance less established than Pgp.
E. Glutathione:
  • Thiol-Based Detoxification: This cellular thiol participates in detoxification pathways, particularly against alkylating agents (e.g., cisplatin) and free radicals (e.g., doxorubicin). Overexpression leads to resistance; glutathione depletion strategies have yielded mixed clinical results.
F. Failure to Engage Apoptosis:
  • Apoptosis Failure: Many cytotoxic drugs induce apoptosis (programmed cell death). Failure to activate apoptosis, often linked to p53 dysfunction ("guardian of the genome"), allows damaged cells to survive and proliferate, leading to resistance. Gene therapy targeting this mechanism is under investigation.
III. Summary and Conclusion
Clinical drug resistance is a complex, multifactorial problem. The relative contribution of each mechanism varies greatly between patients. Further research into cell cycle regulation, cell life, and cell death is essential to overcome this major obstacle in cancer treatment. Understanding these diverse mechanisms is vital for developing strategies to circumvent drug resistance and improve cancer therapy outcomes.



Picture
0 Comments

Oncology-Anti-Microtubule Agents

4/4/2025

0 Comments

 

Oncology-Anti-Microtubule Agents

I. Core Concept:

Anti-microtubule agents, also known as "spindle poisons," are a class of cancer drugs that target tubulin, a protein forming microtubules crucial for cell division. By disrupting microtubule function, these agents prevent cancer cell proliferation. This makes tubulin a key target for anti-cancer drug development.

II. Key Players:

  • Tubulin: The protein building block of microtubules. Mutations affecting tubulin can lead to drug resistance.
  • Microtubules: Cellular structures essential for cell division and other vital processes. Disruption of microtubules leads to cell death.
  • Taxanes (Paclitaxel, Docetaxel): A significant advancement in anti-cancer chemotherapy, showing considerable success in the 1990s. Current research focuses on improving their delivery and mitigating side effects.
  • Vinca Alkaloids (Vincristine, Vinblastine, Vindesine, Vinorelbine): Another class of anti-microtubule agents. They differ in their specific mechanisms and clinical applications.

III. Mechanism of Action:

Anti-microtubule agents work by binding to tubulin, thereby:

  • Destabilizing polymerized tubulin: Preventing the formation of functional microtubules (e.g., Vinca Alkaloids).
  • Stabilizing polymerized tubulin: Preventing microtubule depolymerization, leading to dysfunctional microtubules (e.g., Taxanes).

IV. Clinical Significance

Agent

Mechanism

Indications

Administration (mg/m²)

Main Toxicities

Pharmacokinetics & Metabolism

Clinical Comments

Vincristine (VCR)

Destabilization (β-tubulin)

Leukemias, lymphomas, pediatric tumors, SCLC, myeloma

0.5–1.4 q 1–4wk

Neuropathy

Metabolized in the liver

Induces multi-drug resistance (MDR) via P-glycoprotein (Pgp).

Vinblastine (VBL)

Destabilization (β-tubulin)

Lymphomas, germ cell tumors, KS, breast cancer

6–10 q 2–4wk

Neutropenia, neuropathy

Metabolized in the liver

Vindesine (VDS)

Destabilization (β-tubulin)

NSCLC, breast cancer, prostate, lymphomas

2–4 q 1–3wk

Neutropenia, neuropathy

Metabolized in the liver

Randomized trials showed no advantage over treatments without VDS.

I. Microtubule-Targeting Agents: Mechanisms of Action

These drugs exert their anti-cancer effects by interfering with microtubule dynamics, essential for cell division and function.

  • Paclitaxel (P) and Docetaxel (D): Microtubule Stabilizers: They bind to microtubules, preventing their depolymerization (disassembly). This leads to cell cycle arrest and ultimately apoptosis (programmed cell death). Additional mechanisms include anti-angiogenesis (blocking blood vessel formation to tumors), disruption of Ki-Ras function (a cancer-promoting protein), and apoptosis induction through bcl-2 phosphorylation.
  • Estramustine phosphate (ep): Microtubule Destabilizer: Unlike P and D, ep binds to microtubule-associated proteins, promoting microtubule disassembly. This also disrupts cell division.

II. Drug Specifics

Drug

Mechanism

Useful Indications

Drug Administration (mg/m²)

Main Toxicities

Pharmacokinetics & Metabolism

Clinical Comments

Paclitaxel (P)

Microtubule Stabilizer

Ovarian, breast, lung cancers (others)

135-175 (q 3wk) IV

Neutropenia, Neurotoxicity

Liver metabolized

Toxicities are dose and schedule-dependent. Steroid pre-medication reduces hypersensitivity. Resistance linked to Pgp and β-tubulin. P53 mutations increase sensitivity.

Docetaxel (D)

Microtubule Stabilizer

Breast, lung cancers (others)

100 (q 3wk) IV, 75 (q 3wk if elevated LFTs)

Neutropenia, Fluid Retention Syndrome (FRS)

Liver metabolized

Steroid pre-medication reduces and delays FRS. Tau and β4-tubulin expression correlate with sensitivity.

Estramustine (ep)

Microtubule Destabilizer

Prostate Cancer

560mg x 2/day orally

GI Issues

75% oral absorption, t1/2 20-40h

Primarily subjective responses in prostate cancer. Often combined with other anti-microtubule agents. Resistance potentially linked to β(iii and IVa)-tubulin and tau overexpression.

III. Key Concepts & Considerations:

  • Dose and Schedule Dependency: The toxicity profiles of these drugs are significantly affected by the dosage and administration schedule. Weekly schedules are under investigation for both P and D.
  • Resistance Mechanisms: Resistance to these drugs can develop through various mechanisms, including alterations in the expression or structure of β-tubulin (the protein that forms microtubules), overexpression of proteins like P-glycoprotein (Pgp), which pumps drugs out of cells, and mutations in genes like p53.
  • Combination Therapy: These drugs are often used in combination with other chemotherapeutic agents (e.g., cisplatin, carboplatin, doxorubicin, etoposide) to enhance efficacy and overcome resistance.
  • Pharmacokinetics: Understanding how each drug is metabolized (primarily hepatic for P and D) and its half-life is essential for determining appropriate dosage regimens and managing toxicity.
Picture
0 Comments

Oncology-Topoisomerase Inhibitors

4/4/2025

0 Comments

 

Oncology-Topoisomerase Inhibitors

I. Topoisomerases: The Basics

  • Function: Topoisomerases are nuclear proteins crucial for managing DNA topology (shape and structure). They regulate DNA supercoiling, a process essential for replication, transcription, and recombination.
  • Types: Eukaryotes possess two main types:
    • Topoisomerase I (Topo I): Cleaves and rejoins one strand of DNA, relaxing supercoils.
    • Topoisomerase II (Topo II): Cleaves both strands of DNA, allowing another DNA duplex to pass through the break.

II. Topoisomerase I Inhibitors

  • Camptothecin (CPT) and Derivatives: CPT, derived from the Camptotheca acuminata tree, stabilizes the covalent complex between Topo I and DNA, preventing relegation and leading to DNA damage.
    • Limitations of CPT: High toxicity initially hindered its widespread use.
    • Licensed Derivatives: Irinotecan (CPT-11) and Topotecan are currently used clinically. They differ in administration schedules and toxicity profiles.
  • Side Effects (common to both Irinotecan and Topotecan): Neutropenia, diarrhea (early or late onset), thrombocytopenia, anemia, alopecia, nausea, and vomiting.
  • Clinical Pharmacology:
    • Absorption and Distribution: Both are administered intravenously (IV), and Topotecan can also be absorbed orally (30-50% bioavailability). Both distribute widely throughout the body, including the cerebrospinal fluid (CSF).
    • Metabolism and Excretion: Topotecan undergoes minimal metabolism and is primarily renally excreted. Its clearance is directly related to creatinine clearance. Irinotecan (CPT-11) is a prodrug; it's converted to the active form, SN-38, by carboxylesterases. SN-38 is eliminated via glucuronidation and biliary excretion. Liver dysfunction necessitates dose adjustments.

III. Topoisomerase II Inhibitors

  • Etoposide and Teniposide: These drugs inhibit Topo II's ability to rejoin cleaved DNA, resulting in:
    • High levels of double-stranded DNA breaks.
    • Increased mutations.
    • Illegitimate recombination.
    • Apoptosis (programmed cell death).
  • Pharmaceutical Properties: Poor water solubility necessitates formulation with excipients (e.g., polysorbate for etoposide, Cremophor EL for teniposide). Etoposide can be given orally or IV; teniposide is IV only.
  • Clinical Use: Widely used in adult and pediatric cancers. Etoposide is more commonly used in first-line treatments, especially for small-cell lung cancer (SCLC) and germ cell tumors. Teniposide is not licensed in the UK.
  • Toxicity: Similar toxicity profiles for both, including neutropenia, alopecia, mucositis, infusion-related blood pressure changes, and hypersensitivity reactions.
  • Clinical Pharmacology:
    • Absorption: Etoposide absorption is non-linear, with reduced bioavailability at higher doses.
    • Protein Binding: Both are highly protein-bound; low albumin levels increase free drug concentration and toxicity.
    • Metabolism and Excretion: Both are extensively metabolized, with etoposide exhibiting faster elimination than teniposide. Etoposide clearance shows a linear relationship with creatinine clearance.

IV. Key Differences Summarized:

Feature

Topoisomerase I Inhibitors (e.g., Irinotecan, Topotecan)

Topoisomerase II Inhibitors (e.g., Etoposide, Teniposide)

Target Enzyme

Topoisomerase I

Topoisomerase II

Mechanism

Stabilizes Topo I-DNA complex, preventing relegation

Inhibits DNA relegation, causing double-stranded breaks

Key Side Effects

Neutropenia, Diarrhea

Neutropenia, Alopecia, Mucositis

Metabolism

Topotecan: Renal; Irinotecan: Hepatic (SN-38)

Extensive hepatic metabolism

0 Comments

Oncology-Chemotherapy: Cisplatin and its Analogues

4/4/2025

0 Comments

 

Oncology-Chemotherapy: Cisplatin and its Analogues -

I. Cisplatin

A. Mechanism of Action:

  • Cisplatin directly binds to DNA, forming intra-strand and inter-strand cross-links.
  • This inhibits DNA synthesis by altering the DNA template. It acts as a bifunctional alkylating agent.
  • Cytotoxic effects are cell cycle-independent.
  • Synergistic effects with anti-metabolites are observed (mechanism not fully understood, possibly due to DNA repair malfunction).

B. Side Effects:

  • Highly emetogenic (causes vomiting).
  • Dose-dependent nephrotoxicity (kidney damage).
  • Peripheral neuropathy (nerve damage).
  • Ototoxicity (hearing damage, tinnitus).
  • Can cause anaemia (low red blood cell count).
  • Relatively low toxicity to white blood cells and platelets.

C. Dosage:

  • Standard dose limit: 100 mg/m² as a single daily dose.
  • Higher doses used in clinical trials, often with neuroprotective agents.
  • Alternative schedules exist (e.g., five daily injections of 20 mg/m² for teratoma).
  • Dosage based on empirical body surface area calculations; no clear pharmacokinetic/pharmacodynamic relationship.
  • Clearance is rapid initially, then slower due to plasma protein binding. Prolonged in renal insufficiency.

D. Clinical Indications:

  • Major advancement in testicular cancer treatment (80%+ complete response in metastatic disease).
  • Used in ovarian cancer, genitourinary tumors, squamous carcinomas (head and neck, non-small-cell lung cancer).
  • Frequently used in combination with other cytotoxic agents for various solid cancers and pediatric tumors.

II. Carboplatin

A. Overview:

  • Cisplatin analogue with reduced toxicity compared to cisplatin.
  • Clinically viable alternative to cisplatin in most situations, except for germ cell tumors where cisplatin may be preferred.
  • Cross-resistance with cisplatin: patients resistant to one will likely be resistant to the other.

B. Side Effects:

  • Significant: Thrombocytopenia (low platelet count), leucopenia (low white blood cell count) – both worse around day 14.
  • Less Significant: Renal toxicity, neurological toxicity, otological toxicity, nausea and vomiting (occasional), mild alopecia (hair loss), rare visual disturbances, allergy (2% incidence).

C. Dosage:

  • Initial dosage based on body surface area led to variable thrombocytopenia.
  • Pharmacokinetic-based dosing is the standard: Dose calculated to achieve a specific AUC (area under the curve).
  • Commonly used formula: Dose (mg) ≈ AUC (mg/mL.min) × (GFR + 25)
    • AUC: Typically 4-7 mg/mL.min (varies with administration frequency, prior treatment, combination drugs).
    • GFR: Glomerular filtration rate (mL/min), ideally measured by isotope clearance (e.g., 51CrEDTA), 24-hour urinary creatinine clearance acceptable.

D. Activity:

  • Less toxic substitute for cisplatin with similar indications.
  • Increased thrombocytopenia may be a disadvantage in some combinations.
  • Reduced non-hematological toxicity is advantageous in high-dose regimens with bone marrow/stem cell rescue.

E. Pharmacokinetic Interactions:

  • Unlike cisplatin, carboplatin does not affect hepatic cytochrome P450 enzymes.
  • Pharmacokinetic interactions with other drugs are rare.

III. Oxaliplatin

A. Overview:

  • Platinum analogue differing chemically and possibly mechanistically from cisplatin and carboplatin.
  • Broad spectrum of in vitro activity, differing from cisplatin/carboplatin.
  • Extensively used in colorectal cancer (adjuvant and advanced disease).
  • Broad-spectrum activity, used in other cancers (e.g., upper GI).

B. Dosage:

  • Common regimens:
    • 85 mg/m² every 2 weeks as a 2-6 hour infusion.
    • 130 mg/m² every 3 weeks as a 2-6 hour infusion.
  • Many other dosing regimens exist, including chronomodulated infusion with fluorouracil.

C. Limitation:

  • Cumulative dosage limited by the development of peripheral neuropathy (usually reversible upon drug withdrawal).

IV. Summary Table:

Feature

Cisplatin

Carboplatin

Oxaliplatin

Mechanism

DNA cross-linking

DNA cross-linking

DNA cross-linking (possibly different)

Toxicity

High (nephrotoxicity, neurotoxicity, ototoxicity, emesis)

Lower (thrombocytopenia, leucopenia prominent)

Lower (cumulative peripheral neuropathy)

Dosage

Empirical (BSA)

Pharmacokinetic (AUC-based)

Various regimens (infusion)

Clinical Use

Broad (testicular, ovarian, etc.)

Broad (alternative to cisplatin)

Colorectal, other cancers

Key Advantage

Highly effective

Reduced non-hematological toxicity

Broad spectrum

Key Disadvantage

High toxicity

Increased thrombocytopenia

Cumulative neuropathy

0 Comments

Oncology-Anti-Metabolites

4/4/2025

0 Comments

 
Oncology-Anti-Metabolites
I. Introduction to Anti-Metabolites
  • Mechanism: Anti-metabolites disrupt normal nucleic acid metabolism, primarily during the S-phase of the cell cycle (Fig 5.1). They are widely used cytotoxic agents, not limited to cancer treatment.
II. Anti-Folates
  • Folate Biochemistry: Understanding folate biochemistry is crucial. Thymidylate synthase (TS) is rate-limiting in thymidylate (dTTP) synthesis, converting dUMP to dTTP using CH2-FH4. Dihydrofolate reductase (DHFR) maintains the supply of reduced folate.
III. Methotrexate (MTX)
  • Clinical Use: Widely used in various cancers (breast, osteosarcoma, GI, choriocarcinoma).
  • Pharmacology: Well absorbed orally (<25mg/m²), usually administered IV. Hepatic metabolism to active 7-OH-MTX; 7-10% biliary excretion. Dose adjustments usually unnecessary with hepatic dysfunction. Fluid collections (ascites, pleural effusions) increase toxicity due to reduced clearance. Excretion inhibited by probenecid, penicillins/cephalosporins, NSAIDs.
  • Toxicities: Mucositis, myelosuppression, nephrotoxicity.
IV. Thymidylate Synthase (TS) Inhibitors
  • Direct vs. Indirect Inhibitors: New agents directly inhibit TS (e.g., Raltitrexed), unlike indirect inhibitors (e.g., 5-FU, MTX).
  • Raltitrexed: Prolonged TS inhibition via polyglutamation. Triphasic elimination (renal excretion ~50%). Active in breast and colorectal cancers. Toxicities: myelosuppression, diarrhea, transaminitis. Unpredictable severe toxicities limit widespread use.
V. Fluoropyrimidines
  • Mechanism: Prodrugs activated intracellularly, inhibiting pyrimidine synthesis.
A. Fluorouracil (5-FU)
  • Clinical Use: Widely used, particularly in breast, GI, and head/neck cancers.
  • Mechanism: Metabolized to FdUMP, forming a stable complex with TS in the presence of CH2-FH4, inhibiting TS. Also inhibits RNA synthesis and pre-ribosomal RNA processing.
  • Pharmacology: IV administration (bolus or prolonged infusion). Short initial half-life; hepatic, renal, and lung clearance. Active metabolites (5dUMP, FUTP) have variable pharmacokinetics.
  • Toxicities: Myelosuppression, stomatitis, diarrhea (longer administration). Prolonged infusion minimizes bone marrow effects but causes hand-foot syndrome, neurotoxicity, and cardiotoxicity.
B. Fluorouracil Prodrugs
  • Tegafur (Uftoral®): Oral combination with uracil (1:4 molar ratio). Licensed in many countries (not the US). Active mainly in colorectal and other GI cancers.
  • Capecitabine: Oral prodrug, preferentially activated in tumor and liver tissue. Potential replacement for prolonged 5-FU infusion. Active in various cancers; licensed for breast and GI cancers. Further clinical trials ongoing.
  • Floxuridine: IV administration; metabolized to 5-FU and FdUMP. Primarily used in hepatic artery infusion for colon cancer due to lower toxicity than 5-FU. Not licensed in the UK.
C. Modulation of Fluorouracil
  • Fluorouracil + Folinic Acid (FA): Mainstay colon cancer treatment. FA increases CH2-FH4 supply, potentiating 5-FU's interaction with TS. Higher response rate but increased toxicity compared to 5-FU alone.
VI. Anti-Purines
  • Clinical Use: Treat leukemias, immunosuppressants (azathioprine), antivirals (aciclovir, ganciclovir).
  • Pemetrexed: Novel TS inhibitor; also inhibits DHFR and GARFT. Licensed for mesothelioma and NSCLC. Toxicity reduced by B12 and folate supplementation.
  • 6-Mercaptopurine (6-MP) & 6-Thioguanine (6-TG): Inhibit de novo purine synthesis; nucleotide products incorporated into DNA. Synergistic with MTX. Resistance develops due to HGPRT deficiency. Variable oral bioavailability.
  • Pharmacology: Short half-life; primarily metabolized. 6-MP is a xanthine oxidase substrate (dose adjustments needed with allopurinol). Poor CSF penetration.
  • Toxicities: Myelosuppression; 6-MP can cause hepatotoxicity; nausea, vomiting, mucositis (more common with 6-MP). Main indication: hematological malignancies (6-MP for ALL maintenance, 6-TG for AML).
VII. Cytosine Analogues
A. Cytarabine (Ara-C)
  • Mechanism: Active transport; ara-CTP incorporated into DNA, inhibiting DNA polymerases and possibly phospholipid synthesis. Damaged DNA susceptible to repair.
  • Clinical Use: Active in NHL and AML, not solid tumors. Renal excretion of deaminated compound. Continuous infusion improves activity due to rapid clearance.
  • Toxicities: Emesis, alopecia, myelosuppression, “ara-C syndrome” (fevers, myalgias, rash, keratoconjunctivitis, arthralgias), rarely lung/pancreatic damage.
B. Gemcitabine (dF-CTP)
  • Mechanism: Better membrane permeation and deoxycytidine kinase affinity than ara-C. Prolonged intracellular retention via self-potentiation (bi- and triphosphates facilitate phosphorylation and inhibit catabolism). dF-CTP incorporated into DNA, followed by only one normal nucleotide (“masked termination”), protecting DNA from repair. Schedule-dependent activity (usually weekly IV × 3 out of 4 weeks).
  • Clinical Use: Wide range of cancers, notably pancreatic carcinoma (modest survival advantage).
  • Toxicities: Flu-like symptoms, transaminitis, peripheral edema, myelosuppression, nephrotoxicity. Synergy with cisplatin (schedule-dependent).
VIII. Adenosine Analogues
  • Clinical Use: Low-grade NHL, Waldenström’s macroglobulinemia, CLL. Interact with adenosine deaminase (ADA).
  • Toxicities: Myelosuppression (lymphocytes), reduced NK cell activity.
A. Fludarabine:
  • Resistant to ADA; actively transported, phosphorylated, incorporated into DNA/RNA; possible topo II inhibition. Can cause hemolytic anemia. Useful in CLL.
B. Pentostatin:
  • High ADA affinity; stable complex, resulting in enzyme inhibition. Major indication: hairy cell leukemia. Inhibits DNA synthesis and repair.
C. Cladribine:
  • Resistant to ADA; phosphorylated, incorporated into DNA. Used for hairy cell leukemia.
IX. Hydroxycarbamide
  • Mechanism: Inhibits ribonucleotide reductase, reducing deoxynucleotide availability. Crosses blood-brain barrier.
  • Clinical Use: Myeloproliferative disorders.
  • Toxicities: Myelosuppression, GI toxicities, hyperpigmentation.



Picture
0 Comments

Oncology-Anti-Tumor Antibiotics

4/4/2025

0 Comments

 

Oncology-Anti-Tumor Antibiotics

I. Anthracyclines (Doxorubicin, Daunorubicin, Epirubicin, Idarubicin)

A. Mechanism of Action:

  • Multiple Effects: Anthracyclines affect cell surfaces, signal transduction (activating protein kinase C), and, most importantly, DNA topoisomerase II (topo II). The exact contribution of each effect to cytotoxicity remains unclear.
  • Free Radical Generation: Reduction to highly reactive compounds and free radical production contribute to their anti-cancer activity, but also cause cardiotoxicity (due to lower antioxidant defenses in the heart).
  • Topoisomerase II Inhibition: Anthracyclines bind to the DNA-topo II complex, preventing DNA strand rejoining after temporary breaks, leading to DNA damage and cell death.

B. Drug Resistance:

  • MDR1 Gene/P-glycoprotein (Pgp): This efflux pump, encoded by the MDR1 gene, actively removes anthracyclines from cells, limiting their effectiveness. Manipulating Pgp expression has had limited clinical success.
  • MRP (Multidrug Resistance-associated Protein): Another efflux pump contributing to anthracycline resistance.

C. Pharmacokinetics & Metabolism:

  • Rapid Initial Decline: After IV administration, plasma levels fall rapidly due to tissue distribution and DNA binding.
  • Slow Elimination: Subsequent metabolism and elimination result in slow plasma concentration decline over several days.
  • Liver Function: Dose reduction is recommended for patients with abnormal liver function to minimize toxicity.

D. Clinical Use:

  • Doxorubicin & Epirubicin: IV administration for breast cancer, sarcoma, and hematological malignancies.
  • Daunorubicin & Idarubicin: Primarily used in acute leukemia (idarubicin can be oral).

E. Toxicity:

  • Acute Toxicities (5-10 days post-treatment): Myelosuppression (bone marrow suppression), mucositis (mouth sores), and alopecia (hair loss - reversible). Extravasation (leakage from IV site) is severe and lacks effective treatment.
  • Cumulative Cardiotoxicity: Dose-dependent heart failure due to free radical accumulation. Risk is <5% below 450mg/m² doxorubicin, significantly increasing at higher doses. Pre-existing heart disease and radiation therapy increase risk. Liposomal doxorubicin reduces cardiotoxicity. Epirubicin, daunorubicin, and idarubicin are less cardiotoxic than doxorubicin.

II. Other Anti-Tumor Antibiotics

A. Mitoxantrone:

  • Mechanism: Binds to DNA, interacts with topo II, but produces fewer free radicals than anthracyclines. Also a Pgp substrate.
  • Clinical Use: Less cardiotoxic alternative to doxorubicin in advanced breast cancer, NHL, and non-lymphocytic leukemia, but less effective.

B. Dactinomycin (Actinomycin-D):

  • Mechanism: Strong DNA intercalator, inhibiting RNA and protein synthesis. Pgp substrate.
  • Clinical Use: Highly active against childhood cancers.

C. Mitomycin (MMC):

  • Mechanism: Active against various solid tumors; also used as a radiosensitizer.
  • Clinical Use: Used in combination chemotherapy for breast cancer, NSCLC, GI cancer, and as a radiosensitizer in anal cancer.
  • Toxicity: Delayed and cumulative myelosuppression (especially thrombocytopenia). Administered every 6 weeks due to this toxicity. Uncommon side effects include hemolytic-uremic syndrome, pulmonary fibrosis, and cardiac complications.

III. Key Differences & Summary Table

Drug Class

Example Drugs

Mechanism of Action

Major Toxicity

Clinical Use

Anthracyclines

Doxorubicin, Daunorubicin, Epirubicin, Idarubicin

Topoisomerase II inhibition, free radical generation

Cardiotoxicity, myelosuppression, mucositis

Various cancers, especially breast and leukemia

Mitoxantrone

Mitoxantrone

Topoisomerase II inhibition

Less cardiotoxic than anthracyclines

Breast cancer, NHL, non-lymphocytic leukemia

Dactinomycin

Dactinomycin (Actinomycin-D)

DNA intercalation, inhibits RNA/protein synthesis

Varies

Childhood cancers

Mitomycin (MMC)

Mitomycin

DNA alkylation, radiosensitizer

Delayed, cumulative myelosuppression

Solid tumors, radiosensitization

0 Comments

Oncology-Alkylating Agents

4/4/2025

0 Comments

 

Oncology-Alkylating Agents

I. Introduction

Alkylating agents are antiproliferative cytotoxic drugs used in cancer chemotherapy. They function by covalently binding to DNA via alkyl groups, primarily causing cross-linking. This leads to cell cycle arrest (G1-S transition), followed by either DNA repair or apoptosis (programmed cell death).

II. Clinical Use

Alkylating agents are widely used to treat:

  • Leukemia
  • Lymphoma
  • A range of solid tumors

III. Mechanisms of Resistance

Resistance to alkylating agents is complex and varies depending on the specific agent. Key mechanisms include:

  • Enhanced DNA repair: Increased expression of enzymes like O6-alkyltransferase (especially relevant for nitrosoureas).
  • Increased detoxification: Elevated levels of:
    • Glutathione
    • Metallothionein
    • Glutathione-S-transferase

IV. Examples of Alkylating Agents

This section details key characteristics, uses, and toxicities of specific alkylating agents. Pay close attention to the differences in their mechanisms, side effects, and clinical applications.

Agent

Description

Clinical Use

Key Toxicities

Notes

Melphalan

Nitrogen mustard derivative + phenylalanine (targets dividing cells via amino acid uptake)

Leukemia, lymphoma, solid tumors

Myelosuppression (bone marrow suppression)

Chlorambucil

Phenylbutyric acid derivative of nitrogen mustard (oral administration)

Solid and hematological malignancies

Myelosuppression

Well-absorbed orally

Cyclophosphamide

Extensively used; pro-drug activated by hepatic P450

Broad range of cancers

Marrow suppression, alopecia, nausea, vomiting

Relatively low non-hematological toxicity; used in high-dose regimens

Ifosfamide

Isomer of cyclophosphamide; metabolized to chloroacetaldehyde (toxic metabolite)

Broad range of cancers

Alopecia, hemorrhagic cystitis (mitigated by mesna)

Mesna co-administration is crucial to reduce hemorrhagic cystitis

Busulfan

Specific use in Chronic Myelogenous Leukemia (CML)

CML

Myelosuppression, hepatic veno-occlusive disease, hyperpigmentation, pulmonary fibrosis

Well absorbed from GI tract

Carmustine (BCNU)

Lipophilic molecule

CNS tumors, high-dose conditioning

Myelosuppression

Temozolomide

Newer agent

Glioma, melanoma

Important Note: Both cyclophosphamide and ifosfamide are pro-drugs activated by hepatic cytochrome P450 to form nitrogen mustards. Remember the differences in their toxicity profiles and how those toxicities are managed clinically.

0 Comments

Oncology-Chemotherapy Combination Therapy

4/4/2025

0 Comments

 
Oncology-Chemotherapy Combination Therapy
This guide summarizes the rationale behind using combination chemotherapy to treat cancer.
I. Limitations of Single Cytotoxic Agents:
  • Mechanism: Cytotoxic chemotherapy kills cancer cells by targeting:
    • Nucleic acid chemistry
    • DNA/RNA production
    • Cell division mechanics (e.g., spindle poisons)
  • Lack of Selectivity: A major limitation is the lack of selectivity. Existing drugs damage both cancer and normal dividing cells.
  • Classification of Cytotoxic Agents: They are classified by:
    • Chemical properties/mechanisms of action
    • Source (e.g., natural products)
    • Cell cycle specificity (phase-specific or not)
II. Principles of Combination Chemotherapy Regimen Design:
Effective combination therapy relies on several key principles:
  • Single-Agent Activity: Each drug must effectively kill cancer cells on its own within the specific tumor type.
  • Different Mechanisms of Action: Drugs should target different cellular processes to avoid overlapping effects and resistance.
  • Non-Overlapping Toxicity: Drugs should have different side effect profiles to minimize overall toxicity to the patient, allowing for higher doses.
  • Cell Cycle Diversity: Selecting drugs that target different phases of the cell cycle maximizes the number of cells killed, as tumor cells divide asynchronously.
  • Avoiding Shared Resistance Mechanisms: Drugs should not all be susceptible to the same resistance mechanisms, such as multidrug resistance (MDR).
III. Rationale for Combination Therapy:
  • Increased Fractional Cell Kill: The primary goal is to increase the proportion of cancer cells killed, leading to improved tumor response. Higher doses generally lead to greater cell kill (within limits).
  • Addressing Asynchronous Cell Division: Tumors contain cells in different stages of the cell cycle; combination therapy with drugs acting at different stages increases efficacy.
  • Overcoming Multidrug Resistance (MDR): Some tumors exhibit MDR, often due to efflux pumps that expel drugs. Combination therapy can help circumvent this resistance.
IV. Targeted Therapies and Combination Strategies:
  • Targeted Therapies: These newer therapies specifically inhibit processes crucial to cancer cell survival (e.g., angiogenesis inhibition, EGFR blockade). They often target oncogene function.
  • Integration with Existing Therapies: Much research is ongoing to determine the optimal way to combine targeted therapies with cytotoxic chemotherapy to maximize patient benefit.​
V. Key Terms & Concepts:
  • Cytotoxic Chemotherapy: Treatment that kills cancer cells directly.
  • Selectivity: The ability of a drug to target cancer cells without harming normal cells.
  • Cell Cycle: The series of events leading to cell division.
  • Phase-Specific Drugs: Drugs that are most effective during specific phases of the cell cycle.
  • Multidrug Resistance (MDR): The ability of cancer cells to resist multiple chemotherapy drugs.
  • Targeted Therapies: Drugs that specifically target cancer-related molecules or pathways.
  • Angiogenesis Inhibition: Blocking the formation of new blood vessels that supply the tumor.
  • EGFR Blockade: Inhibition of the Epidermal Growth Factor Receptor, a protein involved in cell growth and division.
Picture
0 Comments

Oncology -Total Body Irradiation (TBI)

4/4/2025

0 Comments

 
Oncology-Total Body Irradiation (TBI)
I. What is TBI?
Total body irradiation (TBI) is a high-dose radiation therapy used to eliminate residual malignant disease and ablate bone marrow before stem cell transplantation. This is often combined with high-dose chemotherapy. The goal is to improve cure rates for sensitive tumors.
II. Aims of TBI:
  • Eliminate residual cancer: Destroy any remaining cancer cells.
  • Ablate bone marrow: Completely destroy the patient's existing bone marrow to allow successful engraftment (establishment) of donor stem cells.
  • Immune suppression: Reduce the immune system's response, especially crucial for non-haplotype identical grafts (where the donor's and recipient's HLA genes aren't completely matched).
III. Indications for High-Dose Therapy (using TBI):
  • Hematological malignancies: Relapsed acute leukemia, high-grade non-Hodgkin lymphoma (NHL), and myeloma often precede bone marrow or stem cell transplants that use TBI.
  • Other malignancies: Neuroblastoma is an example.
IV. Types of Hematopoietic Reconstitution (Bone Marrow Replacement):
  • Autologous: The patient receives their own stem cells (peripheral or cryopreserved marrow) harvested before high-dose therapy.
  • Allogeneic: The patient receives stem cells from a matched or mismatched donor (often a family member or from a donor registry). Mismatched grafts may be one haplotype identical.
V. Pre-Treatment Screening:
Crucial before TBI to assess patient suitability and mitigate risks:
  • Disease remission: The cancer must be in remission (dormant) before proceeding.
  • Organ function: Adequate renal (kidney), cardiac (heart), hepatic (liver), and pulmonary (lung) function is essential to handle treatment toxicity.
  • Medication assessment: Review medication history for potential interactions or side effects that could exacerbate TBI's effects, including:
    • Neurotoxicity: Asparaginase
    • Renal toxicity: Platinum, Ifosfamide
    • Pulmonary toxicity: Methotrexate (MTX), Bleomycin
    • Cardiac toxicity: Cyclophosphamide, Anthracyclines
  • Sanctuary sites: Assess the need for additional treatment to areas where cancer cells may hide (sanctuary sites), such as the central nervous system (CNS), testes, and mediastinum.
VI. Preparation for TBI:
  • Antiemetics: Administer intravenously (IV) to prevent nausea and vomiting. Commonly includes a 5-HT antagonist and dexamethasone.
  • Sedation: May require additional sedation with phenobarbital or diazepam. Ketamine anesthesia may be necessary for very young children.
VII. TBI Technique:
  • Linear accelerator: A linear accelerator is used, optimally with 6MV energy.
  • Fractionated TBI: Preferred for convenience, delivering radiation in smaller doses over several days.
  • Patient positioning: Patient lies on a couch behind a Perspex sheet (for consistent skin dose), alternating between side and back positions. Opposed fields are used for half the treatment time.
  • Distance: The couch is positioned at an extended distance to achieve the necessary body coverage.
  • Dose inhomogeneity: Dose distribution isn't uniform due to body shape and tissue density differences (e.g., lungs). Bolus or lung shielding may compensate, but often isn't necessary with the described schedules.
  • Dose-limiting organ: The lungs are the most vulnerable organ to radiation damage.
VIII. Dose Calculation:
  • Dosimetry: Paired lithium fluoride dose meters or diodes measure the dose distribution throughout the body. Readings are taken from defined sites (lung, mediastinum, abdomen, pelvis).
  • Midline dose: Averaging anterior-posterior (AP) and posterior-anterior (PA) readings.
  • CT scanning: Whole-body CT scans with planning software can also determine doses.
IX. Dose Schedules:
  • Adults: Optimal fractionated doses are 13.2-14.4 Gy (depending on the prescription point). The maximum lung dose (dose-limiting) shouldn't exceed 14.4 Gy.
  • Children: May tolerate slightly higher doses than adults. The MRC protocol uses eight 1.8 Gy fractions over four days. Many other schedules exist.
X. Toxicity of TBI:
A. Acute Effects (occurring shortly after treatment):
  • Nausea and vomiting: Start about 6 hours after the first fraction.
  • Parotid swelling: Occurs in the first 24 hours, resolving spontaneously.
  • Hypotension: Low blood pressure.
  • Fever: Usually controlled with steroids.
  • Diarrhoea: Occurs around day 5 due to gastrointestinal (GI) mucositis (inflammation of the mucous membranes).
B. Delayed Toxicity (occurring weeks to years later):
  • Pneumonitis: Lung inflammation causing shortness of breath and characteristic X-ray changes.
  • Somnolence: Sleepiness, anorexia (loss of appetite), and sometimes nausea (6-8 weeks post-treatment), resolves within 7-10 days.
  • Cataracts: Develop in <20% of patients, increasing in incidence for 2-6 years.
  • Hormonal changes: Azoospermia (sterility in males) and amenorrhea (absence of menstruation) are common; occasional cases of maintained fertility exist.
  • Hypothyroidism: Thyroid damage, sometimes combined with pituitary damage.
  • Growth impairment (children): Impaired growth hormone production and early epiphyseal fusion (bone growth plate closure) lead to stunted growth.
  • Second malignancies: A 5-fold increased risk of developing new cancers, including brain tumors, oral and rectal carcinomas, and lymphoid system malignancies (due to prolonged immune suppression).
Picture
0 Comments
<<Previous

    Kembara Xtra 

    Facts about medicine and its subtopic such as anatomy, physiology, biochemistry, pharmacology, medicine, pediatrics, psychiatry, obstetrics and gynecology and surgery. 

    Picture

    Archives

    April 2025
    March 2025
    February 2025
    January 2025
    December 2024
    November 2024
    October 2024
    September 2024
    August 2024
    June 2024
    May 2024
    April 2024
    March 2024
    February 2024
    January 2024
    December 2023
    August 2023
    July 2023
    June 2023

    Categories

    All
    Clinical Procedures
    Dermatology
    Infectious Diseases And Microbiology
    Medical Physiology
    Medical Science
    Medical Terms
    Medicine
    Pathology
    Pharmacology
    Surgery
    Symptoms And Signs

    RSS Feed

Powered by Create your own unique website with customizable templates.