Antimetabolites & Nucleic Acid Synthesis

Choose a study mode

Play Quiz
Study Flashcards
Spaced Repetition
Chat to Lesson

Podcast

Play an AI-generated podcast conversation about this lesson
Download our mobile app to listen on the go
Get App

Questions and Answers

Why are antimetabolites classified as such in the context of cancer treatment?

  • They accelerate the metabolism of toxic substances in cancer cells.
  • They directly destroy cancerous cells through oxidation.
  • They interfere with metabolic processes crucial for DNA synthesis. (correct)
  • They enhance the body's natural metabolic rate to fight cancer.

During which phase of the cell cycle do most antimetabolite drugs exert their primary cytotoxic effects?

  • G1 phase
  • G2 phase
  • S phase (correct)
  • M phase

A researcher is studying a new drug that inhibits the formation of pyrimidine nucleotides. Which nitrogenous bases would be directly affected by this drug?

  • Cytosine, Thymine, and Uracil (correct)
  • Adenine and Thymine
  • Adenine and Guanine
  • Guanine and Uracil

In a molecular biology experiment, researchers are analyzing a nucleic acid sequence. They identify a nitrogenous base composed of two rings. Which base is most likely present in the sequence?

<p>Guanine (B)</p> Signup and view all the answers

A scientist is designing a drug that specifically targets ribonucleotide reductase. What direct effect would this drug have on nucleotide synthesis?

<p>Prevent the conversion of ribonucleotides to deoxyribonucleotides. (C)</p> Signup and view all the answers

A researcher is investigating a new compound that inhibits the synthesis of thymidine. In which type of nucleic acid would this compound have the most direct impact?

<p>DNA (C)</p> Signup and view all the answers

During DNA replication, a cell is deficient in deoxyribonucleotides. Which of the following enzymes would be most effective in restoring the balance of nucleotides required for replication?

<p>Ribonucleotide Reductase (D)</p> Signup and view all the answers

Suppose a new antimetabolite drug is designed to inhibit the formation of Purine nucleotides. What are the likely targets of this drug?

<p>Adenine and Guanine (D)</p> Signup and view all the answers

Why do drugs that disrupt DNA synthesis commonly cause myelosuppression?

<p>They interfere with the rapid division of bone marrow precursor cells required for producing blood cells. (C)</p> Signup and view all the answers

What is the primary characteristic of megaloblastic anemia caused by DNA synthesis-disrupting drugs?

<p>Increased MCV (mean corpuscular volume) indicating larger than normal red blood cells. (B)</p> Signup and view all the answers

An Absolute Neutrophil Count (ANC) below what value is generally considered neutropenia, indicating a high risk of bacterial infections?

<p>500 (D)</p> Signup and view all the answers

Which of the following is a common finding on a peripheral blood smear in a patient with megaloblastic anemia?

<p>Hypersegmented neutrophils (C)</p> Signup and view all the answers

Why does Cytarabine (Ara-C) inhibit DNA polymerase?

<p>It is structurally similar to cytidine and gets incorporated into DNA, preventing further DNA synthesis. (A)</p> Signup and view all the answers

What is a key difference between Cladribine and other antimetabolite drugs?

<p>Cladribine is cell cycle nonspecific, affecting cells outside of the S-phase. (C)</p> Signup and view all the answers

What is the mechanism of action of Methotrexate?

<p>Inhibits dihydrofolate reductase, reducing the synthesis of tetrahydrofolate. (C)</p> Signup and view all the answers

How does methotrexate indirectly decrease DNA synthesis?

<p>By inhibiting the synthesis of tetrahydrofolate, which is required for nucleotide synthesis (A)</p> Signup and view all the answers

What is the role of N5,N10-tetrahydrofolate (THF) in thymidine synthesis?

<p>It serves as a co-factor for thymidylate synthase in converting deoxyuridine monophosphate to thymidine monophosphate. (A)</p> Signup and view all the answers

Why is methotrexate used in the treatment of autoimmune diseases?

<p>As an immunosuppressant to reduce immune system activity. (D)</p> Signup and view all the answers

How does cytarabine ultimately halt DNA synthesis?

<p>By mimicking cytidine and getting incorporated into DNA, thereby inhibiting DNA polymerase. (D)</p> Signup and view all the answers

A patient is receiving chemotherapy and their Absolute Neutrophil Count (ANC) drops to 400. What is the most significant risk associated with this condition?

<p>Elevated risk of life-threatening bacterial infections (A)</p> Signup and view all the answers

Which of the following antimetabolite drugs is most closely associated with causing neurotoxicity at high doses?

<p>Cytarabine (D)</p> Signup and view all the answers

In the synthesis of thymidine, what specific step is directly dependent on tetrahydrofolate?

<p>Conversion of deoxyuridine monophosphate to thymidine monophosphate. (C)</p> Signup and view all the answers

A patient with rheumatoid arthritis is prescribed methotrexate. What is the primary reason for using methotrexate in this scenario?

<p>To inhibit the production of DNA and RNA in immune cells, reducing inflammation. (D)</p> Signup and view all the answers

Which of the following is the primary reason methotrexate is used in patients with autoimmune diseases?

<p>To eliminate the need for long-term steroid use. (A)</p> Signup and view all the answers

How does leucovorin reverse the myelosuppressive effects of methotrexate?

<p>By converting to tetrahydrofolate without needing dihydrofolate reductase. (A)</p> Signup and view all the answers

Why does methotrexate commonly cause mucositis?

<p>It damages the GI epithelium, leading to loss of mucosal integrity. (D)</p> Signup and view all the answers

Methotrexate-induced lung injury is believed to be what type of reaction?

<p>An allergic reaction. (A)</p> Signup and view all the answers

How does 5-Fluorouracil (5-FU) inhibit cell growth?

<p>By forming a mutant Uridine monophosphate that inhibits thymidylate synthase. (A)</p> Signup and view all the answers

How does leucovorin affect the action of 5-Fluorouracil (5-FU)?

<p>It enhances the effects of 5-FU by increasing the binding of mutant Uridine monophosphate to thymidylate synthase. (C)</p> Signup and view all the answers

Which of the following cancers is 5-FU NOT typically used to treat?

<p>Lung cancer. (A)</p> Signup and view all the answers

Which of the following is a unique side effect associated with 5-FU that is not commonly seen with other chemotherapy drugs?

<p>Cerebellar ataxia. (A)</p> Signup and view all the answers

What is the mechanism by which 6-Mercaptopurine (6-MP) disrupts purine synthesis?

<p>It is converted to a mutant form that inhibits multiple steps in the purine salvage pathway. (C)</p> Signup and view all the answers

Which enzyme is responsible for adding 6-MP to PRPP to form Thioinosinic acid?

<p>HGPRT. (B)</p> Signup and view all the answers

A patient on methotrexate develops severe myelosuppression. Which of the following interventions is most appropriate?

<p>Administer leucovorin to reverse the methotrexate's effects. (A)</p> Signup and view all the answers

A patient is being treated with 5-FU for colorectal cancer. Which of the following side effects would warrant immediate evaluation for potential cardiac complications?

<p>Sudden onset of chest pain and EKG changes. (B)</p> Signup and view all the answers

A patient has been taking low-dose methotrexate for several months to manage rheumatoid arthritis and presents with increasing shortness of breath and a non-productive cough. What is the most likely cause?

<p>Methotrexate-induced lung injury. (C)</p> Signup and view all the answers

A physician is considering using methotrexate for a patient with severe psoriasis who has not responded to topical treatments. Which of the following pre-existing conditions would be a contraindication to using methotrexate?

<p>Active hepatitis. (D)</p> Signup and view all the answers

A patient receiving 6-MP is also prescribed allopurinol for gout. How does allopurinol impact the metabolism of 6-MP, and what adjustment should be made?

<p>Allopurinol decreases the metabolism of 6-MP, requiring a lower dose. (C)</p> Signup and view all the answers

Which of the following best describes the mechanism of action of HGPRT?

<p>Adds a purine base to PRPP to form a nucleotide. (C)</p> Signup and view all the answers

A patient receiving cyclophosphamide develops hyponatremia. What physiological mechanism primarily contributes to this electrolyte imbalance?

<p>Stimulation of ADH release and reduced renal response, leading to water retention. (A)</p> Signup and view all the answers

How does 6-MP exert its effects on cells?

<p>By being converted to a mutant nucleotide that inhibits DNA synthesis. (C)</p> Signup and view all the answers

Ifosfamide's toxicity to proximal tubular cells can result in Fanconi syndrome. What set of clinical findings would be most consistent with this syndrome?

<p>Hypokalemia, metabolic acidosis, and increased urine phosphate. (B)</p> Signup and view all the answers

What is the role of Azathioprine in immunosuppression?

<p>It is a pro-drug that is converted to 6-MP. (A)</p> Signup and view all the answers

Why are nitrosoureas, such as carmustine (BCNU) and lomustine (CCNU), particularly effective in treating brain tumors?

<p>They are highly lipid soluble and easily cross the blood-brain barrier. (B)</p> Signup and view all the answers

Why should allopurinol or febuxostat be used with caution in patients taking azathioprine or 6-MP?

<p>They inhibit xanthine oxidase, which is involved in the metabolism of 6-MP, potentially increasing its toxic effects. (D)</p> Signup and view all the answers

A patient is undergoing myeloablation with high-dose busulfan prior to a bone marrow transplant. What is the most likely immediate hematological consequence of this treatment?

<p>Severe pancytopenia. (C)</p> Signup and view all the answers

A patient with gout is prescribed allopurinol. They are also taking azathioprine for an autoimmune condition. What adjustments, if any, should be made to their medication regimen, and why?

<p>Decrease the dose of azathioprine to prevent toxicity due to reduced metabolism. (D)</p> Signup and view all the answers

How does 6-ThioGuanine differ structurally from guanine, and what is its mechanism of action?

<p>It contains a sulfur atom instead of an oxygen atom and decreases cellular levels of IMP, AMP, and GMP. (D)</p> Signup and view all the answers

A patient on long-term busulfan therapy develops a chronic cough and dyspnea. A chest CT reveals ground glass opacities. What is the most likely underlying pulmonary complication?

<p>Pulmonary fibrosis. (C)</p> Signup and view all the answers

Which mechanism of action is NOT associated with anthracycline-induced cell death?

<p>Directly inhibiting ribosome assembly, preventing protein synthesis (C)</p> Signup and view all the answers

What is the primary mechanism of action of hydroxyurea?

<p>Inhibiting the enzyme ribonucleotide reductase. (D)</p> Signup and view all the answers

What is the role of dacarbazine in the ABVD protocol for Hodgkin lymphoma?

<p>It functions as an alkylating agent. (A)</p> Signup and view all the answers

What is a major advantage of using hydroxyurea compared to some other antimetabolite drugs?

<p>It has good oral bioavailability. (C)</p> Signup and view all the answers

What is the role of topoisomerase II in normal cellular function?

<p>To relieve tangles and supercoils in DNA by cutting and resealing DNA strands. (C)</p> Signup and view all the answers

Which component of the MOPP protocol distinguishes it from the ABVD protocol in the treatment of Hodgkin lymphoma?

<p>The use of a mustard agent (mechlorethamine). (C)</p> Signup and view all the answers

How does anthracycline inhibition of topoisomerase II lead to cell death?

<p>It causes the enzyme to break DNA, but prevents it from resealing it, resulting in DNA damage. (B)</p> Signup and view all the answers

In what conditions is hydroxyurea commonly used, and what is the rationale for its use in sickle cell anemia?

<p>Myeloproliferative disorders and sickle cell anemia; increases fetal hemoglobin levels. (A)</p> Signup and view all the answers

A patient receiving ifosfamide is being monitored for potential neurological side effects. Which of the following is a common initial sign of ifosfamide-induced encephalopathy?

<p>Confusion. (A)</p> Signup and view all the answers

A researcher is investigating new treatments for glioblastoma multiforme. Which property of the nitrosourea class of drugs makes them a relevant candidate for this research?

<p>Their lipophilicity, allowing them to cross the blood-brain barrier effectively. (C)</p> Signup and view all the answers

Intercalation of anthracyclines within DNA directly inhibits which processes?

<p>Replication and transcription of DNA strands (C)</p> Signup and view all the answers

Which of the following best describes the mechanism by which alkylating agents affect DNA?

<p>They add an alkyl group to a nucleotide base in DNA. (D)</p> Signup and view all the answers

The generation of free radicals by anthracyclines involves the conversion of a quinone to a semiquinone. What is required for this process?

<p>NADPH and oxygen (B)</p> Signup and view all the answers

A patient undergoing bone marrow transplant preparation with busulfan develops significant hyperpigmentation of the skin. What is the most likely mechanism behind this side effect?

<p>Disruption of normal melanin regulation due to cellular toxicity. (B)</p> Signup and view all the answers

Why is the N7 nitrogen of guanine a common target for alkylating agents?

<p>It is the most exposed and reactive site on guanine. (A)</p> Signup and view all the answers

Which of the following best describes the role of iron in anthracycline-induced free radical formation?

<p>Iron catalyzes the conversion of hydrogen peroxide to hydroxyl radicals, leading to DNA oxidative damage. (B)</p> Signup and view all the answers

A researcher is studying the mechanism by which cyclophosphamide causes SIADH. Which of the following represents the current understanding of this process?

<p>Increased ADH release coupled with decreased renal responsiveness. (D)</p> Signup and view all the answers

A researcher is studying the effects of a novel alkylating agent on cancer cells. Which of the following findings would provide the strongest evidence that the agent is acting as a typical alkylating agent?

<p>Detection of alkylated guanine bases in cellular DNA. (D)</p> Signup and view all the answers

Which of the following is a major clinical concern associated with doxorubicin (Adriamycin) treatment?

<p>Cardiotoxicity potentially leading to systolic heart failure. (D)</p> Signup and view all the answers

A patient is prescribed ifosfamide as part of their chemotherapy regimen. What strategies can be implemented to reduce the risk of nephrotoxicity associated with ifosfamide treatment?

<p>Ensuring adequate hydration and monitoring electrolyte levels. (A)</p> Signup and view all the answers

Which of the following is NOT a typical use for Azathioprine and 6-MP?

<p>Chemotherapy agents. (B)</p> Signup and view all the answers

A patient on 6-MP develops myelosuppression. Which of the following factors is LEAST likely to contribute to this adverse effect?

<p>Increased intake of dietary purines. (D)</p> Signup and view all the answers

A patient is receiving carmustine (BCNU) as part of a high-dose chemotherapy regimen in preparation for a bone marrow transplant. What neurological complications should the medical team be vigilant for?

<p>Encephalopathy and seizures. (A)</p> Signup and view all the answers

What is believed to be the primary mechanism behind anthracycline-induced cardiotoxicity?

<p>Free radical damage to myocytes leading to necrosis. (D)</p> Signup and view all the answers

Why is echocardiogram used in patients treated with doxorubicin?

<p>As a screening tool to detect cardiotoxicity early. (B)</p> Signup and view all the answers

A researcher aims to develop a novel drug that selectively targets tumor cells while minimizing pulmonary toxicity. Which alkylating agent should be avoided as a prototype due to its known association with pulmonary fibrosis?

<p>Busulfan. (D)</p> Signup and view all the answers

A patient with polycythemia vera is being treated with hydroxyurea. Routine blood tests reveal an increase in fetal hemoglobin (HbF) levels. How should the physician interpret this finding?

<p>The increase in HbF may provide additional benefit, especially if the patient also has sickle cell trait. (B)</p> Signup and view all the answers

What is the mechanism of action of dexrazoxane in preventing cardiotoxicity associated with anthracyclines?

<p>It acts as an iron-chelating agent, limiting anthracycline-induced cardiotoxicity. (A)</p> Signup and view all the answers

A physician is selecting a chemotherapy regimen for a patient with Hodgkin lymphoma and is considering both ABVD and MOPP protocols. What factor might lead the physician to favor ABVD over MOPP?

<p>Reduced risk of long-term side effects such as secondary malignancies. (C)</p> Signup and view all the answers

Why is the use of dexrazoxane limited to special circumstances, despite its ability to prevent cardiotoxicity?

<p>It has shown to interfere with benefit of chemotherapy agents. (B)</p> Signup and view all the answers

What is the primary mechanism of action of dactinomycin?

<p>Intercalation into DNA and inhibition of RNA synthesis. (B)</p> Signup and view all the answers

What is the most significant adverse effect associated with dactinomycin?

<p>Myelosuppression (D)</p> Signup and view all the answers

Which of the following characteristics distinguishes bleomycin from other antitumor antibiotics?

<p>It is cell cycle-specific for the G2 phase. (B)</p> Signup and view all the answers

Bleomycin's mechanism of action is most similar to which other antitumor antibiotic?

<p>Doxorubicin (D)</p> Signup and view all the answers

Alkylating agents like nitrogen mustards lead to cell death via what mechanism?

<p>Cross-linking DNA strands, inhibiting replication and causing DNA damage. (D)</p> Signup and view all the answers

Why are patients treated with alkylating agents at risk of developing therapy-related myeloid neoplasms (t-MNs)?

<p>Alkylating agents induce mutations and DNA damage in myeloid cells. (B)</p> Signup and view all the answers

A patient who was treated with cyclophosphamide five years ago is now presenting with fatigue, unexplained bruising, and frequent infections. Which of the following secondary malignancies is most concerning?

<p>Therapy-related acute myeloid leukemia (t-AML) (C)</p> Signup and view all the answers

Which of the following is a common mechanism shared by alkylating agents, topoisomerase II inhibitors, and radiation therapy that leads to therapy-related myeloid neoplasms?

<p>DNA damage and induction of mutations. (D)</p> Signup and view all the answers

Which structural feature is characteristic of nitrogen mustard alkylating agents?

<p>A central nitrogen atom with two chloroethyl groups (D)</p> Signup and view all the answers

Which of the following nitrogen mustards does NOT contain a central nitrogen atom but still functions similarly?

<p>Ifosfamide (D)</p> Signup and view all the answers

Cyclophosphamide is a prodrug that requires bioactivation in the liver. What is the primary enzyme system involved in this activation?

<p>Cytochrome P450 (CYP) system (D)</p> Signup and view all the answers

Phosphoramide mustard is the active cytotoxic metabolite of cyclophosphamide. What is another significant metabolite generated during cyclophosphamide metabolism that contributes to its side effects?

<p>Acrolein (C)</p> Signup and view all the answers

Which of the following mechanisms explains why cyclophosphamide can cause myelosuppression?

<p>It inhibits DNA replication in rapidly dividing bone marrow cells. (B)</p> Signup and view all the answers

A patient on cyclophosphamide develops hematuria and dysuria. Which of the following conditions is most likely causing these symptoms?

<p>Hemorrhagic cystitis (A)</p> Signup and view all the answers

Which strategy is most effective in reducing the risk of hemorrhagic cystitis in patients receiving cyclophosphamide?

<p>Co-administration of mesna (A)</p> Signup and view all the answers

Which strategy is LEAST likely to mitigate nephrotoxicity associated with Cisplatin administration?

<p>Restricting fluid intake to minimize diuresis. (D)</p> Signup and view all the answers

What is the mechanism of action of mesna in preventing hemorrhagic cystitis caused by cyclophosphamide?

<p>It binds to and inactivates acrolein in the urine. (C)</p> Signup and view all the answers

A patient receiving Cisplatin develops acute kidney injury. Which lab findings would MOST likely be associated with this condition?

<p>Increased BUN and creatinine levels. (A)</p> Signup and view all the answers

A patient with severe rheumatoid arthritis is prescribed low-dose oral cyclophosphamide. What is the most likely reason for using cyclophosphamide in this scenario?

<p>To suppress the immune system and reduce autoimmune activity. (A)</p> Signup and view all the answers

How do topoisomerase inhibitors induce cell death in cancer cells?

<p>By breaking DNA strands without allowing resealing. (A)</p> Signup and view all the answers

A patient is receiving cyclophosphamide as part of their chemotherapy regimen. Which of the following would be the MOST appropriate counseling point regarding potential drug interactions?

<p>&quot;Avoid grapefruit juice, as it can alter the metabolism of this medication.&quot; (D)</p> Signup and view all the answers

A researcher is studying the effects of a new drug that inhibits a specific enzyme in the liver. When co-administered with cyclophosphamide, the researcher observes significantly reduced levels of phosphoramide mustard and increased levels of the parent drug. Which enzyme is most likely being inhibited by the new drug?

<p>Cytochrome P450 (CYP) (C)</p> Signup and view all the answers

During which phases of the cell cycle do topoisomerase inhibitors typically exert their primary cytotoxic effects?

<p>S and G2 phases. (A)</p> Signup and view all the answers

What is the MOST significant mechanism of action of Irinotecan and Topotecan?

<p>Inhibition of topoisomerase I. (B)</p> Signup and view all the answers

Which of the following is NOT a typical side effect associated with Irinotecan and Topotecan?

<p>Peripheral neuropathy. (B)</p> Signup and view all the answers

Etoposide and Teniposide are derived from which naturally occurring substance?

<p>Podophyllotoxins. (B)</p> Signup and view all the answers

Which of the following cancers is LEAST likely to be treated with Etoposide or Teniposide?

<p>Brain cancer. (C)</p> Signup and view all the answers

Which feature distinguishes monoclonal antibodies from traditional chemotherapy drugs?

<p>Monoclonal antibodies target specific antigens, whereas chemotherapy drugs are less targeted. (B)</p> Signup and view all the answers

What is the typical route of administration for monoclonal antibodies in cancer treatment?

<p>Intravenous infusion. (A)</p> Signup and view all the answers

What physiological response is MOST directly associated with the occurrence of infusion reactions following monoclonal antibody administration?

<p>Cytokine release. (A)</p> Signup and view all the answers

What is the primary mechanism by which Taxol resistance develops in tumor cells?

<p>Increased tumor cell production of P-glycoprotein. (C)</p> Signup and view all the answers

Why is pre-medication, such as with glucocorticoids and antihistamines typically administered before Taxol?

<p>To prevent hypersensitivity reactions. (D)</p> Signup and view all the answers

Premedication with antihistamines or steroids is MOST likely considered for monoclonal antibodies known to cause:

<p>Severe infusion reactions. (A)</p> Signup and view all the answers

Which characteristic of nab-paclitaxel (Abraxane) reduces the risk of hypersensitivity reactions compared to traditional Taxols?

<p>It is bound to albumin. (D)</p> Signup and view all the answers

Bevacizumab's mechanism of action primarily involves targeting which specific molecule?

<p>VEGF-A. (A)</p> Signup and view all the answers

Why do Taxols commonly induce neuropathy as a side effect?

<p>They interfere with microtubule function, disrupting axonal transport. (C)</p> Signup and view all the answers

What is the primary role of VEGF-A in tumor development and progression?

<p>Stimulation of angiogenesis. (D)</p> Signup and view all the answers

Besides cancer, in which other medical condition are VEGF inhibitors used therapeutically?

<p>Retinopathy. (C)</p> Signup and view all the answers

How do Vinca Alkaloids, such as Vincristine and Vinblastine, disrupt the cell cycle?

<p>By inhibiting the formation of the mitotic spindle, arresting cells in metaphase. (A)</p> Signup and view all the answers

A patient undergoing chemotherapy develops burning parasthesias in their fingers and toes. Which drug class is most likely responsible for these symptoms?

<p>Vinca Alkaloids (D)</p> Signup and view all the answers

What is the mechanism of action of platinum-based drugs like Cisplatin?

<p>Cross-linking DNA strands. (B)</p> Signup and view all the answers

Which of the following side effects is most commonly associated with platinum agents such as Cisplatin and Carboplatin?

<p>Peripheral neuropathy (D)</p> Signup and view all the answers

A patient receiving chemotherapy develops hearing loss. Which class of chemotherapy drugs is most likely responsible?

<p>Platinum agents (B)</p> Signup and view all the answers

Why are platinum agents classified as cell cycle nonspecific?

<p>They can kill cancer cells in any phase of the cell cycle. (A)</p> Signup and view all the answers

A researcher aims to develop a drug that enhances the effect of Taxols. Which of the following strategies would be most effective?

<p>An agent that inhibits P-glycoprotein. (B)</p> Signup and view all the answers

A patient is prescribed the ABVD protocol for Hodgkin lymphoma. Which drug in this protocol is a Vinca Alkaloid?

<p>Vinblastine (D)</p> Signup and view all the answers

Given the mechanism of action of Taxols, which cellular process would be directly affected?

<p>Microtubule dynamics during mitosis (C)</p> Signup and view all the answers

Which patient is most likely to experience peripheral neuropathy as a result of their chemotherapy treatment?

<p>All of the above (D)</p> Signup and view all the answers

A patient is prescribed a chemotherapy regimen. The doctor is concerned about potential neurotoxicity. Which of the following drugs should the doctor be most cautious about in terms of dosing?

<p>Vincristine (D)</p> Signup and view all the answers

Which of the following explains why bleomycin's toxicity primarily affects the skin and lungs?

<p>The enzyme bleomycin hydrolase, which inactivates bleomycin, is present in lower levels in the skin and lungs. (B)</p> Signup and view all the answers

A patient undergoing chemotherapy with bleomycin develops red, dark streaks on their skin. This is most likely indicative of which dermatological side effect?

<p>Flagellate erythema (B)</p> Signup and view all the answers

Which of the following is the dose-limiting adverse effect most commonly associated with bleomycin?

<p>Pulmonary toxicity (B)</p> Signup and view all the answers

A 70-year-old patient with a history of chronic obstructive pulmonary disease (COPD) is started on bleomycin for cancer treatment. Which of the following is the MOST important consideration regarding the potential for adverse effects?

<p>The patient's age and pre-existing pulmonary disease increase the risk of pulmonary toxicity. (C)</p> Signup and view all the answers

What is the fundamental building block of a microtubule?

<p>Tubulin (B)</p> Signup and view all the answers

Why are microtubules particularly important for neuronal function?

<p>They facilitate the transport of substances along the axon. (C)</p> Signup and view all the answers

During which phase of the cell cycle do microtubule inhibitors exert their primary effects?

<p>M phase (A)</p> Signup and view all the answers

Which phase of mitosis is characterized by the alignment of chromosomes along the metaphase plate?

<p>Metaphase (B)</p> Signup and view all the answers

How do Taxol drugs such as paclitaxel and docetaxel affect microtubule function?

<p>They enhance tubulin polymerization, leading to microtubules that cannot break down. (B)</p> Signup and view all the answers

How does the mechanism of action of Taxol drugs lead to cell cycle arrest?

<p>By blocking the transition from metaphase to anaphase. (A)</p> Signup and view all the answers

Which of the following is the MOST accurate description of alkaloids?

<p>Naturally occurring, nitrogen-containing bases often derived from plants. (B)</p> Signup and view all the answers

Which cellular process relies heavily on the dynamic growth and collapse of microtubules?

<p>Cellular transport (C)</p> Signup and view all the answers

If a researcher wants to study the effects of a drug that prevents chromosome separation during cell division, which phase of mitosis should they focus on?

<p>Metaphase/Anaphase transition (D)</p> Signup and view all the answers

A new drug is discovered that binds to tubulin and prevents its polymerization. What direct effect would this drug have on mitosis?

<p>Inhibition of mitotic spindle formation (C)</p> Signup and view all the answers

A scientist is developing a drug that targets the microtubules in cancer cells. Which of the following characteristics would make the drug MOST effective as a chemotherapy agent?

<p>Specifically targets and disrupts the function of microtubules only in rapidly dividing cells. (B)</p> Signup and view all the answers

A patient with a gastrointestinal stromal tumor (GIST) is being treated with Imatinib. What is the primary mechanism of action of Imatinib in this context?

<p>Inhibiting the tyrosine kinase activity of the c-KIT receptor. (C)</p> Signup and view all the answers

A patient on Imatinib develops periorbital edema. What is the most likely mechanism causing this side effect?

<p>Fluid retention caused by altered ion transport. (B)</p> Signup and view all the answers

Rituximab is being considered for a patient with severe rheumatoid arthritis. What is the primary target of Rituximab that makes it useful in treating this condition?

<p>B-cells expressing the CD20 surface marker. (A)</p> Signup and view all the answers

A patient receiving Rituximab develops new neurological symptoms. Which opportunistic infection should be of greatest concern?

<p>Progressive multifocal leukoencephalopathy (PML). (B)</p> Signup and view all the answers

Why is it important to screen patients for Hepatitis B before initiating Rituximab therapy?

<p>Rituximab can lead to reactivation of latent Hepatitis B infection. (C)</p> Signup and view all the answers

A postmenopausal woman with ER-positive breast cancer is being treated with Tamoxifen. What is the rationale for using Tamoxifen in this case?

<p>It competitively binds to estrogen receptors in breast cancer cells, blocking estrogen's effects. (B)</p> Signup and view all the answers

A woman taking Tamoxifen is advised about the drug's potential side effects. Which of the following is a known risk associated with Tamoxifen use?

<p>Increased risk of endometrial cancer. (D)</p> Signup and view all the answers

Why does Tamoxifen increase the risk of deep vein thrombosis (DVT) and pulmonary embolism (PE)?

<p>It has estrogen-agonizing effects that lead to an increase in clotting factors. (D)</p> Signup and view all the answers

Bevacizumab is known to cause cardiovascular side effects due to its inhibition of the VEGF system. What is the primary mechanism by which this inhibition leads to hypertension?

<p>Vasoconstriction due to reduced nitric oxide-mediated vasodilation. (B)</p> Signup and view all the answers

How does Raloxifene's mechanism of action differ from Tamoxifen's in the treatment of breast cancer?

<p>Raloxifene has anti-estrogen effects in both the breast and the uterus, whereas Tamoxifen has estrogen agonist effects on the uterus. (A)</p> Signup and view all the answers

A patient on Bevacizumab therapy is scheduled for elective surgery. Considering the drug's mechanism of action, what is the most significant risk associated with this patient undergoing surgery?

<p>Delayed wound healing due to VEGF's role in maintaining blood vessel health. (A)</p> Signup and view all the answers

Raloxifene is primarily used to treat osteoporosis in postmenopausal women due to which specific effect?

<p>Its estrogen-agonistic effect on bone. (D)</p> Signup and view all the answers

Cetuximab is prescribed for a patient with colon cancer. What is the mechanism of action of Cetuximab?

<p>It binds to the extracellular domain of EGFR, preventing EGF binding and receptor activation. (A)</p> Signup and view all the answers

A patient with a known c-KIT mutation is diagnosed with a gastrointestinal stromal tumor (GIST). What is the significance of the c-KIT mutation in the tumor's development?

<p>It causes a gain-of-function mutation, leading to constitutive activation of the receptor and uncontrolled cell growth (C)</p> Signup and view all the answers

A patient receiving Cetuximab develops an acneiform rash on their face. What is the most accurate statement regarding this adverse effect?

<p>It is thought to correlate positively with the drug's efficacy in some patients. (C)</p> Signup and view all the answers

A researcher is investigating a new monoclonal antibody for treating B-cell malignancies. To which cell surface marker would this antibody likely bind to induce B-cell depletion?

<p>CD20 (B)</p> Signup and view all the answers

A patient on Tamoxifen experiences hot flashes. What is the underlying mechanism causing this side effect?

<p>Blockage of estrogen receptors, mimicking menopausal symptoms. (C)</p> Signup and view all the answers

A colorectal cancer patient with a known K-ras mutation is being considered for targeted therapy. Which of the following statements best describes how the K-ras mutation affects the response to Cetuximab?

<p>The mutation results in constitutive activation of downstream signaling, circumventing EGFR inhibition by Cetuximab. (C)</p> Signup and view all the answers

A researcher is investigating potential drug targets in cancer cells and discovers a novel protein that, when mutated, leads to continuous activation of cell growth pathways, independent of EGFR. Which of the following proteins is analogous to this discovery?

<p>K-ras (C)</p> Signup and view all the answers

A patient is switched from Tamoxifen to Raloxifene for breast cancer prevention. What is the primary reason for this change, considering their effects on the uterus?

<p>Raloxifene has anti-estrogenic effects on the uterus, reducing the risk of endometrial cancer. (A)</p> Signup and view all the answers

Which of the following best describes the function of the KIT protein when it binds to the KIT ligand?

<p>Stimulation of cell growth (D)</p> Signup and view all the answers

Erlotinib is prescribed for a patient with non-small cell lung cancer, but unlike Cetuximab, it is administered orally. What is the primary mechanism of action that differentiates Erlotinib from Cetuximab?

<p>Erlotinib directly inhibits the tyrosine kinase activity of EGFR. (B)</p> Signup and view all the answers

A patient on Erlotinib for lung cancer develops a significant acne-like rash. How should the physician interpret this adverse effect?

<p>Potentially as an indicator of the drug's efficacy, suggesting a possible benefit from the treatment. (A)</p> Signup and view all the answers

Why are aromatase inhibitors primarily used in postmenopausal women with estrogen receptor-positive breast cancer?

<p>Premenopausal women produce estrogen primarily in the ovaries, rendering aromatase inhibitors less effective. (B)</p> Signup and view all the answers

A patient with non-small cell lung cancer is being treated with Erlotinib, an EGFR tyrosine kinase inhibitor. What is the expected outcome of Erlotinib's mechanism of action on the EGFR?

<p>Inhibition of tyrosine kinase activity, reducing downstream signaling. (A)</p> Signup and view all the answers

Aromatase inhibitors prevent the production of estrogen by blocking which enzymatic conversion?

<p>Conversion of androstenedione to estrone and testosterone to estradiol. (C)</p> Signup and view all the answers

Imatinib is used in the treatment of chronic myeloid leukemia (CML). What is the specific molecular target of Imatinib in CML cells?

<p>A BCR-ABL fusion protein with tyrosine kinase activity. (C)</p> Signup and view all the answers

A patient undergoing treatment with Imatinib for chronic myeloid leukemia (CML) asks how the drug works at the molecular level. Which of the following explanations is most accurate?

<p>It inhibits the tyrosine kinase activity of the BCR-ABL protein, preventing downstream signaling. (C)</p> Signup and view all the answers

Which of the following is a significant adverse effect associated with the use of aromatase inhibitors?

<p>Osteoporosis and increased risk of bone fracture. (A)</p> Signup and view all the answers

Besides CML, Imatinib is also used to treat what other type of tumor?

<p>Gastrointestinal stromal tumors (GISTs) (C)</p> Signup and view all the answers

Trastuzumab is a monoclonal antibody that targets which receptor?

<p>Human epidermal growth factor receptor 2 (HER2). (A)</p> Signup and view all the answers

A researcher is investigating new compounds to treat CML and wants to develop a drug similar to Imatinib but with potentially broader efficacy. What molecular target should this new drug primarily inhibit?

<p>The tyrosine kinase activity of the BCR-ABL fusion protein. (D)</p> Signup and view all the answers

What is the primary mechanism by which Trastuzumab is believed to inhibit tumor growth, besides direct inhibition of proliferation?

<p>Stimulating antibody-dependent cell-mediated cytotoxicity (ADCC). (C)</p> Signup and view all the answers

Which of the following best describes antibody-dependent cell-mediated cytotoxicity (ADCC)?

<p>A mechanism where a cell is tagged by antibodies and killed by effector cells. (B)</p> Signup and view all the answers

A patient with chronic myeloid leukemia (CML) who initially responded well to Imatinib begins to show signs of resistance. Which of the following mechanisms is most likely contributing to this resistance?

<p>Mutation in the BCR-ABL kinase domain, reducing Imatinib's binding affinity. (B)</p> Signup and view all the answers

Besides Imatinib, which other tyrosine kinase inhibitors are also used in the treatment of CML?

<p>Dasatinib and nilotinib (B)</p> Signup and view all the answers

What is a notable adverse effect associated with Trastuzumab, and how does it typically manifest?

<p>Cardiac toxicity, often manifesting as an asymptomatic fall in left ventricular ejection fraction. (B)</p> Signup and view all the answers

How does Trastuzumab-induced cardiomyopathy differ from anthracycline-induced cardiomyopathy?

<p>Trastuzumab cardiotoxicity is often reversible upon discontinuation of the drug and is not dose-dependent. (D)</p> Signup and view all the answers

If a patient develops cardiomyopathy while on Trastuzumab and subsequently recovers left ventricular ejection fraction, what is a possible treatment strategy?

<p>Rechallenge the patient with Trastuzumab, as it is sometimes well-tolerated after recovery. (A)</p> Signup and view all the answers

What cellular component is common among all members of the human epithelial receptor (HER) family?

<p>An inner tyrosine kinase domain. (C)</p> Signup and view all the answers

In the context of cancer treatment, what is the primary goal of using aromatase inhibitors in postmenopausal women with estrogen receptor-positive breast cancer?

<p>To block the stimulation of breast cancer cells by estrogen, thereby slowing or stopping their growth. (C)</p> Signup and view all the answers

Of the aromatase inhibitors mentioned, which one possesses a steroid chemical structure?

<p>Exemestane. (C)</p> Signup and view all the answers

Which of the following is another common name used to refer to the HER2 receptor?

<p>ERB2. (A)</p> Signup and view all the answers

If a researcher is investigating the effects of Trastuzumab on a patient's immune response, which type of immune cell would be of particular interest due to its role in ADCC?

<p>Natural killer (NK) cells. (A)</p> Signup and view all the answers

A patient undergoing Trastuzumab treatment experiences dyspnea, fatigue, and edema. Which of the following is the most likely cause?

<p>Heart failure secondary to Trastuzumab-induced cardiomyopathy. (B)</p> Signup and view all the answers

Flashcards

Antimetabolites

Chemotherapy drugs that block the formation of DNA components, inhibiting DNA synthesis.

S phase

The phase of the cell cycle when DNA is synthesized; the primary target of antimetabolites.

Pyrimidines

Nitrogenous bases with a single-ring structure, including Cytosine, Thymine, and Uracil.

Purines

Nitrogenous bases with a double-ring structure, including Adenine and Guanine.

Signup and view all the flashcards

Nucleotide

A nitrogenous base attached to a ribose sugar and a phosphate group.

Signup and view all the flashcards

Pyrimidine Nucleotides

Nucleotides containing the pyrimidine bases (Cytosine, Thymine, Uracil).

Signup and view all the flashcards

Purine Nucleotides

Nucleotides containing the purine bases (Adenine and Guanine).

Signup and view all the flashcards

Ribonucleotide Reductase

Enzyme that converts ribonucleotides to deoxyribonucleotides by removing a hydroxyl group.

Signup and view all the flashcards

Myelosuppression

Suppression of bone marrow activity, leading to reduced production of blood cells.

Signup and view all the flashcards

Megaloblastic Anemia

A type of anemia characterized by larger than normal red blood cells (high MCV) and often hypersegmented neutrophils.

Signup and view all the flashcards

Neutropenia

An abnormally low count of neutrophils in the blood.

Signup and view all the flashcards

Cytarabine (Ara-C)

A chemotherapy drug that is a pyrimidine analog, mimicking cytidine and inhibiting DNA polymerase.

Signup and view all the flashcards

Cytarabine's Mechanism

Inhibition of DNA polymerase, preventing DNA synthesis.

Signup and view all the flashcards

Cladribine

A purine analog that mimics adenosine and is highly toxic to lymphocytes.

Signup and view all the flashcards

Cladribine Cell Cycle

Effective even when cells are not in S-phase

Signup and view all the flashcards

Methotrexate

A drug that mimics folate and inhibits dihydrofolate reductase, thus decreasing DNA synthesis.

Signup and view all the flashcards

Dihydrofolate Reductase

An enzyme important for folate metabolism, converts dihydrofolate to tetrahydrofolate.

Signup and view all the flashcards

N5,N10-Tetrahydrofolate

A cofactor required by thymidylate synthase to convert deoxyuridine monophosphate to thymidine monophosphate which is needed for DNA synthesis.

Signup and view all the flashcards

Thymidylate Synthase

Enzyme which converts Deoxyuridine monophosphate to Thymidine monophosphate

Signup and view all the flashcards

Methotrexate MOA

Blocks the synthesis of tetrahydrofolate, which is required for synthesis of DNA, RNA and some proteins.

Signup and view all the flashcards

Thymidine

A nucleotide necessary for DNA synthesis.

Signup and view all the flashcards

Steroid Sparing Agent

Agents that can reduce the amount of steroids needed for a patient

Signup and view all the flashcards

Hairy Cell Leukemia

A rare form of leukemia.

Signup and view all the flashcards

Leucovorin

Drug used to reverse myelosuppression caused by methotrexate.

Signup and view all the flashcards

Mucositis

Mouth soreness, a common side effect of methotrexate and other chemotherapy agents.

Signup and view all the flashcards

Methotrexate-Induced Lung Injury

Rare lung injury caused by methotrexate, potentially leading to pulmonary fibrosis.

Signup and view all the flashcards

5-Fluorouracil (5-FU)

Antimetabolite drug that mimics uracil.

Signup and view all the flashcards

5-Fluoro Deoxyuridine Monophosphate

5-FU is converted to this, inhibiting thymidylate synthase.

Signup and view all the flashcards

Thymineless Death

Cell death due to the inability to synthesize thymidine, caused by 5-FU.

Signup and view all the flashcards

Leucovorin with 5-FU

In contrast to methotrexate, it enhances the effects of 5-FU.

Signup and view all the flashcards

Colorectal Cancer

A major cancer type treated with 5-FU.

Signup and view all the flashcards

Cerebellar Ataxia (5-FU)

Rare CNS side effect of 5-FU, affecting coordination.

Signup and view all the flashcards

Coronary Vasospasm (5-FU)

Cardiac side effect of 5-FU, leading to chest pain.

Signup and view all the flashcards

6-Mercaptopurine (6-MP)

Drug that mimics purine bases.

Signup and view all the flashcards

HGPRT

Enzyme that adds 6-MP to PRPP, forming Thioinosinic acid.

Signup and view all the flashcards

HGPRT Function

Adds a base to PRPP, creating nucleotide monophosphates (GMP or IMP).

Signup and view all the flashcards

6-MP Mechanism

Mimics hypoxanthine or guanine, creating a mutant nucleotide that inhibits DNA synthesis.

Signup and view all the flashcards

Azathioprine

A pro-drug converted to 6-MP in the body to suppress the immune system.

Signup and view all the flashcards

Azathioprine/6-MP Uses

Immunosuppressants used to wean patients off steroids, preventing organ rejection and treating autoimmune diseases.

Signup and view all the flashcards

Azathioprine/6-MP Adverse Effects

Myelosuppression, abnormal LFTs, and GI upset.

Signup and view all the flashcards

Xanthine Oxidase Function

Converts xanthine to uric acid.

Signup and view all the flashcards

Xanthine Oxidase Inhibitors

Allopurinol and febuxostat.

Signup and view all the flashcards

Allopurinol/Febuxostat Interaction with Aza/6-MP

It will blunt the breakdown of Azathioprine or 6-MP, increasing the risk of adverse effects.

Signup and view all the flashcards

6-Thioguanine Mechanism

It decreases cellular levels of IMP, AMP, and GMP, which inhibits DNA synthesis.

Signup and view all the flashcards

Hydroxyurea Mechanism

Inhibits ribonucleotide reductase, blocking formation of deoxyribonucleotides and DNA synthesis.

Signup and view all the flashcards

Hydroxyurea Adverse Effect

Myelosuppression.

Signup and view all the flashcards

Hydroxyurea Uses

Myeloproliferative disorders and sickle cell anemia.

Signup and view all the flashcards

Hydroxyurea in Sickle Cell Anemia

Increases fetal hemoglobin levels.

Signup and view all the flashcards

Alkylating Agents

Drugs that add an alkyl group to a nucleotide base in DNA, usually at the N7 nitrogen of guanine.

Signup and view all the flashcards

Alkyl Groups

Molecular groups with the formula Cn H2n+1.

Signup and view all the flashcards

Cyclophosphamide & SIADH

Cyclophosphamide can cause SIADH due to its antidiuretic effects, leading to water retention and hyponatremia.

Signup and view all the flashcards

Ifosfamide & Cystitis

Ifosfamide, similar to cyclophosphamide, can cause hemorrhagic cystitis.

Signup and view all the flashcards

Ifosfamide & Fanconi Syndrome

Ifosfamide can be toxic to proximal tubular cells, potentially causing Fanconi syndrome.

Signup and view all the flashcards

Ifosfamide & Encephalopathy

Ifosfamide can cause encephalopathy presenting as confusion in some patients.

Signup and view all the flashcards

Nitrosoureas: Key Features

Nitrosoureas are bioactivated in the liver and are highly lipid soluble, allowing them to cross the blood-brain barrier.

Signup and view all the flashcards

Nitrosoureas & Brain Tumors

Nitrosoureas (e.g., carmustine, lomustine) are used to treat brain tumors due to their ability to cross the blood-brain barrier.

Signup and view all the flashcards

Busulfan's Primary Use

Busulfan is used for myeloablation in bone marrow transplant protocols to cause severe pancytopenia.

Signup and view all the flashcards

Busulfan & Skin Changes

Busulfan can cause skin changes, including hyperpigmentation.

Signup and view all the flashcards

Dacarbazine: Use

Dacarbazine is an alkylating agent and part of the ABVD protocol for Hodgkin lymphoma.

Signup and view all the flashcards

Procarbazine: Use

Procarbazine is an alkylating agent and part of the MOPP protocol for Hodgkin lymphoma.

Signup and view all the flashcards

Origin of Anti-tumor Antibiotics

Antitumor antibiotics are derived from Streptomyces bacteria.

Signup and view all the flashcards

Key Anthracyclines

Daunorubicin and doxorubicin are important drugs in the anthracycline class.

Signup and view all the flashcards

Busulfan & Pulmonary Toxicity

Busulfan can cause pulmonary toxicity, leading to cough, dyspnea, and potentially pulmonary fibrosis.

Signup and view all the flashcards

Nitrosoureas: Side Effects

Nitrosoureas can cause myelosuppression, pulmonary fibrosis (rare), interstitial nephritis, encephalopathy, and seizures.

Signup and view all the flashcards

Hyperpigmentation

Busulfan can lead to skin hyperpigmentation.

Signup and view all the flashcards

Bleomycin Hydrolase

An enzyme that inactivates bleomycin, with lower levels in the skin and lungs.

Signup and view all the flashcards

Flagellate Erythema

A rare skin condition characterized by red, dark streaks, seen with bleomycin.

Signup and view all the flashcards

Bleomycin-induced Pneumonitis

Inflammation of the lungs, a dose-limiting adverse effect of bleomycin.

Signup and view all the flashcards

Microtubule

Structural element of a cell made of alpha and beta tubulin.

Signup and view all the flashcards

Alpha and Beta Tubulin

Repeating protein units (red and brown units) that form microtubules.

Signup and view all the flashcards

Mitosis

Cellular division of chromosomes

Signup and view all the flashcards

Mitotic Spindle

Structure that separates chromosomes during mitosis.

Signup and view all the flashcards

Metaphase

Phase where chromosomes align on the metaphase plate.

Signup and view all the flashcards

Anaphase

Phase where chromosomes separate and move to opposite ends of the cell.

Signup and view all the flashcards

Cell Cycle Specific (M phase)

Drugs affecting cells only during the mitosis (M) phase.

Signup and view all the flashcards

Alkaloids

Naturally occurring nitrogen-containing bases, derived from plants.

Signup and view all the flashcards

Taxols

Paclitaxel and docetaxel: drugs derived from yew trees.

Signup and view all the flashcards

Taxols Mechanism

Enhance tubulin polymerization, preventing microtubule breakdown.

Signup and view all the flashcards

Taxols Effect on Cell Cycle

Blocks cell division by arresting cells at metaphase.

Signup and view all the flashcards

Paclitaxel and Docetaxel

Alkaloids derived from Yew trees used as mitotic spindle poisons.

Signup and view all the flashcards

Therapy-Related Myeloid Neoplasms (t-MNs)

DNA damage caused by alkylating agents can lead to these malignancies affecting the myeloid cells in bone marrow, often years after treatment.

Signup and view all the flashcards

Nitrogen Mustards

Alkylating agents with a structure similar to mustard gas.

Signup and view all the flashcards

Cyclophosphamide

A common nitrogen mustard alkylating agent, available in IV and oral forms, and bioactivated by the liver.

Signup and view all the flashcards

Liver's Role in Cyclophosphamide Activation

Cyclophosphamide must be activated by this organ to exert its cytotoxic effects.

Signup and view all the flashcards

Phosphoramide Mustard

The cytotoxic active form of cyclophosphamide.

Signup and view all the flashcards

Acrolein

A metabolite of cyclophosphamide that is toxic to the bladder and can cause hemorrhagic cystitis.

Signup and view all the flashcards

Hemorrhagic Cystitis

A condition caused by acrolein toxicity in the bladder, leading to hematuria and dysuria.

Signup and view all the flashcards

Mesna

A drug co-administered with cyclophosphamide to bind and inactivate acrolein in the urine, reducing the risk of hemorrhagic cystitis.

Signup and view all the flashcards

DNA Cross-Linking

Cross-linking of these strands inhibits replication, causes damage, and leads to cell death.

Signup and view all the flashcards

Guanosine

When guanine is part of a nucleotide, it's called...

Signup and view all the flashcards

Long-Term Follow-Up After Alkylating Agent Treatment

Due to the risk of therapy-related myeloid neoplasms (t-MNs), patients should be monitored for...

Signup and view all the flashcards

Nitrogen Mustard Structure

A central nitrogen and two chlorine atoms are key features

Signup and view all the flashcards

Pro-drug Activation

Cyclophosphamide requires this for activation

Signup and view all the flashcards

P450 System Interactions with Cyclophosphamide

Affecting this system can alter cyclophosphamide's activation and toxicity.

Signup and view all the flashcards

P-glycoprotein

A protein that pumps foreign substances, like Taxols, out of cells, contributing to multidrug resistance.

Signup and view all the flashcards

Taxol Hypersensitivity Reactions

Symptoms include dyspnea, wheezing, urticaria, and hypotension.

Signup and view all the flashcards

Nab-paclitaxel (Abraxane)

Albumin-bound paclitaxel, a Taxol variant, has a lower risk of hypersensitivity reactions; premedication is not needed.

Signup and view all the flashcards

Taxol Myelosuppression

Inhibition of bone marrow cell division by Taxols.

Signup and view all the flashcards

Taxol Neuropathy

Burning paresthesias of hands or feet due to microtubule interference.

Signup and view all the flashcards

Vinca Alkaloids

Bind to beta tubulin and inhibit polymerization, preventing mitotic spindle formation and arresting cells in metaphase.

Signup and view all the flashcards

ABVD protocol

A chemotherapy protocol for Hodgkin lymphoma including Adriamycin, Bleomycin, Vinblastine, and Decarbazine.

Signup and view all the flashcards

Vincristine Neurotoxicity

Sensory and motor neuropathy caused by axonal transport loss, presenting as paresthesias or distal weakness.

Signup and view all the flashcards

DNA Drugs

Drugs that disrupt the function of DNA to kill tumor cells.

Signup and view all the flashcards

Platinum Agents

Drugs including Cisplatin, Carboplatin, and Oxaliplatin that cross-link DNA, similar to alkylating agents.

Signup and view all the flashcards

Platinum Agents Mechanism

Cross-link DNA at the N7 nitrogen of guanine.

Signup and view all the flashcards

Platinum Agents Neuropathy

Peripheral sensory neuropathy presenting as pain, burning, or tingling in the feet or hands.

Signup and view all the flashcards

Platinum Agents Ototoxicity

Hearing loss caused by platinum agents.

Signup and view all the flashcards

Platinum Agents GI Distress

Nausea and vomiting caused by platinum agents in up to 90% of patients.

Signup and view all the flashcards

Anthracyclines

A class of antitumor antibiotics with a four-ring structure, including doxorubicin and daunorubicin.

Signup and view all the flashcards

Topoisomerase II

Enzyme that relieves DNA tangles by cutting and resealing DNA strands.

Signup and view all the flashcards

DNA Intercalation

Binding to DNA by inserting between base pairs, blocking DNA and RNA synthesis.

Signup and view all the flashcards

Free Radicals

Unstable molecules that can damage cells; anthracyclines generate these.

Signup and view all the flashcards

Quinone

The portion of anthracyclines that is converted to a semiquinone, leading to free radical production.

Signup and view all the flashcards

Doxorubicin (Adriamycin)

Commonly used anthracycline, used in breast cancer, sarcomas and lymphomas.

Signup and view all the flashcards

Cardiotoxicity

Damage to the heart muscle, a potential side effect of anthracyclines.

Signup and view all the flashcards

Dexrazoxane

Iron chelating agent used to prevent anthracycline-induced cardiotoxicity.

Signup and view all the flashcards

Dactinomycin (Actinomycin D)

Antitumor antibiotic that intercalates in DNA and inhibits RNA synthesis.

Signup and view all the flashcards

Bleomycin

Antitumor antibiotic that binds to DNA and generates free radicals.

Signup and view all the flashcards

Free Radical Generation

Requires NADPH, oxygen, and iron.

Signup and view all the flashcards

Bleomycin Use

Used in lymphomas, germ cell tumors, and head and neck cancer

Signup and view all the flashcards

Topoisomerase Inhibition

Breaks DNA, but doesn't reseal it.

Signup and view all the flashcards

Cisplatin Nephrotoxicity

The primary dose-limiting side effect of Cisplatin, often presenting as acute kidney injury.

Signup and view all the flashcards

Preventing Nephrotoxicity

Administering IV fluids (normal saline) with Cisplatin to increase urine output and reduce kidney exposure to the drug.

Signup and view all the flashcards

Amifostine

A free radical scavenger used to limit nephrotoxicity from Cisplatin, particularly in ovarian cancer patients.

Signup and view all the flashcards

Topoisomerase Inhibitors

Chemotherapy drugs that cut and reseal DNA to relieve tangles and supercoils during DNA replication.

Signup and view all the flashcards

Mechanism of Topoisomerase Inhibitors

The result of topoisomerase inhibitors preventing DNA from resealing, leading to DNA damage and cell death.

Signup and view all the flashcards

Irinotecan & Topotecan

Chemotherapy drugs that inhibit topoisomerase one (breaks single strands).

Signup and view all the flashcards

Side Effects of Irinotecan/Topotecan

Severe diarrhea and myelosuppression.

Signup and view all the flashcards

Etoposide & Teniposide

Chemotherapy drugs that inhibit Topoisomerase II (breaks double strands).

Signup and view all the flashcards

Side Effects of Etoposide/Teniposide

Myelosuppression and nausea/vomiting.

Signup and view all the flashcards

Monoclonal Antibodies

Laboratory-produced antibodies designed to bind to specific antigens, often on cancer cells.

Signup and view all the flashcards

Infusion Reactions

Mild to moderate symptoms like fever, chills, flushing, itching, skin rashes, nausea, vomiting, or diarrhea related to antibody binding.

Signup and view all the flashcards

Bevacizumab (Avastin)

A monoclonal antibody that prevents VEGF-A from binding to VEGF receptors used in solid tumors.

Signup and view all the flashcards

VEGF

Vascular Endothelial Growth Factor: A family of signal proteins that stimulate angiogenesis.

Signup and view all the flashcards

Ranibizumab

A VEGF inhibitor used to prevent blood vessel growth in the retina.

Signup and view all the flashcards

Epidermal Growth Factor (EGF)

A molecule that stimulates cell growth and differentiation by binding to EGFR.

Signup and view all the flashcards

Epidermal Growth Factor Receptor (EGFR)

Tyrosine kinase receptor that binds to EGF, leading to cell growth and differentiation.

Signup and view all the flashcards

Cetuximab

An antibody that binds to the extracellular domain of EGFR, blocking EGF binding and preventing activation of the EGF system.

Signup and view all the flashcards

Acneiform Rash

Common side effect of drugs that interfere with the EGF system.

Signup and view all the flashcards

K-ras

A G-protein downstream of EGFR; activating mutations can cause Cetuximab resistance.

Signup and view all the flashcards

Erlotinib

Oral EGFR tyrosine kinase inhibitor that disrupts the EGFR system and benefits some malignancies.

Signup and view all the flashcards

Non-Small Cell Lung Cancer

Cancer where Erlotinib is frequently used.

Signup and view all the flashcards

Tyrosine Kinase Inhibitor

Inhibits tyrosine kinase activity of the EGFR system, disrupting cell signaling pathways.

Signup and view all the flashcards

Imatinib

Tyrosine kinase inhibitor used in chronic myeloid leukemia (CML).

Signup and view all the flashcards

Philadelphia Chromosome

Chromosome abnormality that leads to the BCR-ABL fusion gene in CML.

Signup and view all the flashcards

BCR-ABL Fusion Gene

A fusion gene produced as a result of the Philadelphia chromosome which leads to a tyrosine kinase protein.

Signup and view all the flashcards

Dasatinib & Nilotinib

Tyrosine kinase inhibitors also used in CML treatment.

Signup and view all the flashcards

Gastrointestinal Stromal Tumors (GIST)

A very rare tumor of the stomach and small intestine, which can be treated by Imatinib.

Signup and view all the flashcards

Vasoconstriction

Inhibition of VEGF causes this condition.

Signup and view all the flashcards

Hypertension

Cardiovascular side effect common in patients receiving Bevacizumab that inhibits VEGF.

Signup and view all the flashcards

Aromatase Inhibitors

Drugs that block estrogen stimulation of breast cancer cells, used in postmenopausal women with estrogen receptor-positive breast cancers.

Signup and view all the flashcards

Examples of Aromatase Inhibitors

Anastrozole, Letrozole, and Exemestane; block estrogen production.

Signup and view all the flashcards

Aromatase

Enzyme that produces estrogens by converting Androstenedione to estrone, and testosterone into estradiol.

Signup and view all the flashcards

Osteoporosis (with Aromatase Inhibitors)

A major adverse effect of aromatase inhibitors due to decreased estrogen.

Signup and view all the flashcards

Trastuzumab (Herceptin)

A monoclonal antibody that targets the HER-2 receptor.

Signup and view all the flashcards

HER-2 Receptor

A surface receptor; its activation leads to cell growth and proliferation, overexpressed in some cancers.

Signup and view all the flashcards

Other names for HER-2

HER2/neu, ERB2, CD340

Signup and view all the flashcards

Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC)

The process of killing cells by the immune system, triggered by antibodies.

Signup and view all the flashcards

Effector Cells in ADCC

Natural killer cells and other immune cells.

Signup and view all the flashcards

Trastuzumab's Mechanism of Action

Trastuzumab works by triggering antibody-dependent cell-mediated cytotoxicity.

Signup and view all the flashcards

Cardiac Toxicity (Trastuzumab)

A potential side effect of Trastuzumab that can lead to a fall in left ventricular ejection fraction or heart failure.

Signup and view all the flashcards

Cause of Trastuzumab Cardiac Toxicity

Blockade of HER2 in myocytes, leading to stunning of the myocardium.

Signup and view all the flashcards

Characteristics of Trastuzumab Cardiotoxicity

Not dose-dependent and often reversible when the drug is discontinued.

Signup and view all the flashcards

Anthracycline Cardiotoxicity

Cardiac toxicity associated with anthracyclines; involves necrosis of the myocardium and is often permanent.

Signup and view all the flashcards

Rechallenge Trastuzumab

Stopping and potentially reintroducing the drug. Only possible if their ejection fraction recovers.

Signup and view all the flashcards

KIT Gene Mutations

Mutations affect the KIT protein (c-KIT or CD 117), a tyrosine kinase receptor on cell surfaces.

Signup and view all the flashcards

Proto-oncogene

A gene that can become an oncogene when mutated.

Signup and view all the flashcards

Imatinib Adverse Effects

Fluid retention, skin rash, rarely pulmonary edema.

Signup and view all the flashcards

Rituximab Mechanism

Binds to the CD20 surface marker on B-cells, leading to the depletion of B-cells.

Signup and view all the flashcards

Rituximab Uses

Non-Hodgkin lymphoma, CLL, rheumatoid arthritis, ITP.

Signup and view all the flashcards

Rituximab Toxicity

Infections like pneumocystis pneumonia, cryptococcal meningitis, PML, and hepatitis B reactivation.

Signup and view all the flashcards

Progressive multifocal leukoencephalopathy (PML)

Caused by reactivation of the JC virus.

Signup and view all the flashcards

Tamoxifen

Selective Estrogen Receptor Modulator; competitive antagonist of estrogen receptor in breast cells.

Signup and view all the flashcards

Tamoxifen Uses

Used in ER-positive breast cancers as primary therapy and for prevention.

Signup and view all the flashcards

Tamoxifen Effects

Estrogen agonist in bone and uterus; increases bone density and endometrial growth.

Signup and view all the flashcards

Tamoxifen Toxicity

Hot flashes, increased risk of DVT/PE, endometrial proliferation, and increased risk of endometrial cancer.

Signup and view all the flashcards

Raloxifene

SERM used for osteoporosis; anti-estrogen in breast and uterus; estrogen actions on bone.

Signup and view all the flashcards

Study Notes

Antimetabolites Overview

  • Antimetabolites are chemotherapy drugs that block the formation of DNA components, inhibiting DNA synthesis, thus treating malignancy.
  • They generally target specific phases of the cell cycle, with a few exceptions.
  • Most of these drugs exhibit toxic effects during the S phase, which is when DNA is synthesized.

Biochemistry Review

  • DNA and RNA contain nitrogenous bases categorized as pyrimidines (single-ring) and purines (double-ring).
  • Pyrimidines: Cytosine, Thymine (DNA only), and Uracil (RNA only)
  • Purines: Adenine and Guanine
  • A nitrogenous base attached to a ribose sugar and a phosphate group becomes a nucleotide.
  • Pyrimidine nucleotides contain pyrimidine bases, and purine nucleotides contain purine bases.
  • Examples include Cytidine (Cytosine), Thymidine (Thymine), Uridine (Uracil), Adenosine, and Guanosine.
  • A ribonucleotide has a hydroxyl group on the sugar molecule and is used in RNA.
  • A deoxyribonucleotide lacks a hydroxyl group in that position and is used in DNA.
  • Deoxyribonucleotides are synthesized by ribonucleotide reductase removing the hydroxyl group from ribonucleotides.

Common Side Effects

  • These drugs disrupt DNA synthesis, targeting rapidly dividing cells like bone marrow precursor cells.
  • Myelosuppression is a common side effect, leading to megaloblastic anemia, thrombocytopenia, and leukopenia
  • Absolute Neutrophil Count (ANC) is monitored; levels below 500 indicate neutropenia, increasing infection risk.
  • Megaloblastic anemia is characterized by reduced hematocrit, increased MCV (large cells), and hypersegmented neutrophils.
  • Defective DNA production, such as from B12 or folate deficiency, or chemotherapy drugs like Methotrexate, 5-FU, or Hydroxyurea, can cause it.

Cytarabine (Ara-C)

  • Cytarabine is a pyrimidine analog with a similar structure to Deoxycytidine, but with a hydroxyl group in an abnormal position.
  • It inhibits DNA polymerase by mimicking Cytidine, preventing DNA polymerase from functioning.
  • Effective in treating leukemia and lymphomas.
  • Adverse effects include myelosuppression, nausea, vomiting, and potential neurotoxicity (peripheral neuropathy, confusion, cerebellar ataxia) at high dosages.

Cladribine

  • Cladribine is a purine analog that mimics adenosine.
  • Highly toxic to lymphocytes, and a drug of choice for hairy cell leukemia.
  • Has effects outside the S-phase and is cell cycle nonspecific.
  • Main adverse effect is myelosuppression.

Methotrexate

  • Methotrexate mimics the structure of folate and indirectly decreases DNA synthesis.
  • It inhibits dihydrofolate reductase, which is important for folate metabolism, leading to blocked synthesis of tetrahydrofolate.
  • Tetrahydrofolate is required for DNA, RNA, and protein synthesis.
  • It blocks the formation of thymidine, which is needed for DNA synthesis.
  • It has clinical use as infusion for malignancies (solid tumors, leukemia, lymphomas) and oral form for immunosuppression in autoimmune diseases.
  • It can be used to wean patients off steroids.
  • It induces pregnancy abortion, mostly in ectopic and tubal pregnancies.
  • A major side effect is myelosuppression, which can be reversed by leucovorin (folinic acid).
  • Leucovorin is converted to tetrahydrofolate without dihydrofolate reductase.
  • Mouth sores are a common adverse effect, damaging GI epithelium leading to pain and bacterial growth also known as mucositis.
  • Additional adverse effects include hepatitis, abnormal LFTs, GI upset.
  • Rarely causes methotrexate-induced lung injury, potentially progressing to pulmonary fibrosis if untreated.

5-Fluorouracil (5-FU)

  • 5-FU mimics the structure of uracil.
  • Converted to five fluoro deoxyuridine monophosphate, inhibiting thymidylate synthase and blocking thymidine formation.
  • Cells undergo a "thymineless death".
  • Leucovorin enhances the effects of 5-FU making the process go faster.
  • Used in colorectal, breast, and pancreatic cancer, and topically for basal cell skin cancer.
  • Adverse effects: myelosuppression, nausea, vomiting, diarrhea, mucositis, CNS effects (cerebellar ataxia, encephalopathy), and coronary vasospasm.

6-Mercaptopurine (6-MP)

  • 6-MP mimics the structure of hypoxanthine and guanine.
  • Added to PRPP by HGPRT.
  • Forms Thioinosinic acid thus inhibiting purine salvage pathway and decreasing cellular levels of IMP, AMP, and GMP.

Azathioprine

  • Azathioprine is a pro-drug converted to 6-MP, exerting the same effects on cells.
  • Used for immunosuppression (steroid-sparing) in inflammatory bowel disease, transplant rejection prevention, and autoimmune diseases.
  • Major adverse effects include myelosuppression, abnormal LFTs, and GI upset.
  • Metabolized by xanthine oxidase; inhibition by allopurinol or febuxostat can increase drug adverse effects.

6-Thioguanine

  • 6-Thioguanine reduces cellular levels of IMP, AMP, and GMP, inhibiting DNA synthesis.
  • It mimics the bases, hypoxanthine, and Guanine.

Hydroxyurea

  • Hydroxyurea inhibits ribonucleotide reductase, blocking the formation of deoxyribonucleotides.
  • Advantage: good oral bioavailability.
  • Major adverse effect: myelosuppression.
  • Used in myeloproliferative disorders and sickle cell anemia to increase fetal hemoglobin levels (mechanism unclear).

Summary of Antimetabolites

  • All drugs inhibit DNA synthesis through different mechanisms.
    • Methotrexate: Folate mimic
    • Drugs mimicking purines (Adenine, Guanine)
    • Drugs mimicking pyrimidines (Cytosine, Thymine, Uracil)
    • Hydroxyurea: Unique mechanism

Alkylating Agents Overview

  • Alkylating agents add alkyl groups to nucleotide bases in DNA, commonly the N7 nitrogen of guanine, leading to DNA cross-linking, inhibited replication, damage, and cell death.
  • These agents are cell cycle nonspecific.

Guanine and Alkylation

  • Guanine's chemical structure has the N7 nitrogen as the site for alkyl group addition.
  • Alkylating agents alkylate two guanine bases and link them together, cross-linking DNA strands.
  • Alkylating agents can cause t-MNs, malignancies of bone marrow myeloid cells, occurring years after exposure.
  • Often presents as acute myeloid leukemia (t-AML) or myelodysplastic syndromes (t-MDS).
  • DNA damage in myeloid cells leads to these secondary cancers.
  • Topoisomerase II inhibitors and radiation therapy can also cause t-MNs.

Nitrogen Mustards

  • Nitrogen mustards are alkylating agents similar to mustard gas.
  • Contain a central nitrogen and two chlorine atoms.
  • Examples: Mechlorethamine (simplest), Cyclophosphamide (very common), Melphalan, Chlorambucil, Ifosfamide (slightly different structure).

Cyclophosphamide

  • Cyclophosphamide available in IV and oral forms.
  • It’s bioavailability is good making it a powerful immunosuppressant in autoimmune diseases.
  • Also used intravenously in solid tumors, lymphomas, and leukemias.
  • A pro-drug bioactivated by the liver via the P450 system into phosphoramide mustard.
  • Metabolized into acrolein, responsible for many side effects.
  • Side effects include myelosuppression and hemorrhagic cystitis caused by acrolein (reduced by hydration and mesna).
  • Can cause SIADH (antidiuretic effects), leading to hyponatremia.

Ifosfamide

  • Ifosfamide is an isomer of cyclophosphamide used in germ cell cancers and sarcomas.
  • Can cause hemorrhagic cystitis.
  • Special side effects include nephrotoxicity (Fanconi syndrome) and encephalopathy.

Nitrosoureas

  • Nitrosoureas contain a nitroso group and urea moiety.
  • Examples: Carmustine (BCNU), Lomustine (CCNU), Streptozotocin, Semustine.
  • Bioactivated in the liver.
  • Highly lipid-soluble and easily cross the blood-brain barrier, so they are often used in brain tumors.
  • Side effects include myelosuppression, rare pulmonary fibrosis, chronic interstitial nephritis (renal failure), encephalopathy, and seizures (especially with high dosages of BCNU).

Busulfan

  • Busulfan is used for myeloablation in bone marrow transplant protocols and for CML treatment.
  • Causes severe pancytopenia, preparing patients for stem cell transplants.
  • Side effects include myelosuppression, skin changes (hyperpigmentation), seizures (high dosages), and pulmonary toxicity (cough, dyspnea, pulmonary fibrosis).

Dacarbazine and Procarbazine

  • Dacarbazine is part of the ABVD protocol for Hodgkin lymphoma (Adriamycin, Bleomycin, Vinblastine, Dacarbazine).
  • Procarbazine is part of the MOPP protocol for Hodgkin lymphoma (Mechlorethamine, Oncovin, Procarbazine, Prednisone).

Antitumor Antibiotics Overview

  • Antitumor antibiotics are naturally occurring drugs derived from Streptomyces bacteria strains.
  • Key drugs: anthracyclines, dactinomycin, and bleomycin.

Anthracyclines

  • Examples: Daunorubicin, Doxorubicin (Adriamycin), Idarubicin, Epirubicin, Mitoxantrone.
  • All have a four-ring structure.
  • Multiple toxic mechanisms:
    • Inhibit topoisomerase II (DNA breaks)
    • Intercalate into DNA (blocks DNA and RNA synthesis)
    • Generate free radicals
  • Cell cycle non-specific.
  • Doxorubicin (Adriamycin) is used in breast cancer, solid tumors, childhood cancers (neuroblastoma, Ewing's sarcoma, osteosarcoma), leukemia, and lymphoma.
  • Cardiotoxicity (systolic heart failure) is a major problem due to free radical damage to myocytes.
  • Early detection through echocardiogram monitoring. _ Cardiotoxicity management is an entire field called cardio oncology.
  • Total dosage limited.
  • Dexrazoxane (iron chelating agent) can prevent cardiotoxicity, but interferes with chemotherapy benefits.

Dactinomycin (Actinomycin D)

  • Multiple mechanisms: intercalates in DNA, inhibits RNA synthesis and transcription, induces double-stranded breaks.
  • Commonly used in childhood cancers (neuroblastoma, Ewing's sarcoma, osteosarcoma).
  • Major adverse effect: myelosuppression.

Bleomycin

  • Binds to DNA and generates free radicals using oxygen and iron.
  • Leads to single and double-stranded DNA breaks.
  • Cell cycle-specific (G2 phase).
  • Used in lymphomas, germ cell tumors, head and neck cancer, squamous cell carcinoma of skin, and cancers of cervix and vulva.
  • Toxicity: inactivated by bleomycin hydrolase (lower levels in skin and lungs, leading to skin and lung toxicity).
  • Skin changes including flagellate erythema.
  • Pulmonary toxicity (dose-limiting adverse effect): pneumonitis (cough, dyspnea, crackles, infiltrates on chest x-ray).
  • More common in older patients with prior pulmonary disease.

Microtubule Inhibitors Overview

  • Microtubules are structural elements in cells and polymers made of repeating alpha and beta tubulin units.
  • Important for flagella, cilia, cellular transport, and mitosis.

Mitosis and Microtubules

  • Mitosis is part of the cell cycle where chromosomes are separated for division.
  • Depends on microtubules to develop the mitotic spindle.
  • Mitotic spindle microtubules attach to chromosomes and pull them apart.
  • Key phases of mitosis are metaphase (chromosomes align on metaphase plate) and anaphase (chromosomes separate).
  • Microtubule inhibitors are cell cycle-specific, functioning only in the M phase (mitosis).
  • Two classes of microtubule inhibitors: Taxols and Vinca Alkaloids.

Taxols

  • Alkaloids are naturally occurring nitrogen-containing bases found in plants or trees.
  • Examples: Paclitaxel and Docetaxel (from yew trees).
  • Mitotic spindle poisons enhance tubulin polymerization, leading to microtubules that cannot break down.
  • Blocks cell cycle at metaphase-anaphase transition.
  • Specific target: beta tubulin.
  • Tumor resistance mechanisms: altered beta tubulin, increased P-glycoprotein production (multidrug resistance protein).
  • Used on solid tumors: ovarian, breast, lung, head and neck, prostate, and bladder cancer.

Adverse Effects of Taxols

  • Hypersensitivity reactions (dyspnea, wheezing, urticaria, hypotension).
  • Pre-medication with glucocorticoids or antihistamines.
  • Nabpaclitaxel (Abraxane): albumin-bound paclitaxel with lower risk of hypersensitivity.
  • Myelosuppression and neuropathy (burning parasthesias of hands or feet).

Vinca Alkaloids

  • Drugs: Vincristine and Vinblastine (from periwinkle plant).
  • Bind to beta tubulin and inhibit polymerization.
  • Prevents mitotic spindle formation, arresting cells in metaphase.
  • Used in breast cancer, germ cell cancers, lymphomas, and ABVD protocol for Hodgkin lymphoma.
  • Adverse effects: myelosuppression, rare SIADH, neurotoxicity (especially Vincristine).
  • Vincristine: dose-limiting toxicity due to axonal transport loss (sensory and motor neuropathy, parasthesias, pain, distal weakness).

DNA Drugs Overview

  • Chemotherapy drugs disrupt DNA function to kill tumor cells
    • Platinum agents and drugs inhibiting topoisomerase one or two.

Platinum Agents

  • Platinum agents include Cisplatin, Carboplatin, and Oxaliplatin.
  • Cross-link DNA (like alkylating agents), commonly at the N7 nitrogen of guanine.
  • Called "alkylating-like" and are cell cycle nonspecific
  • Used in solid tumors: small cell and non-small cell lung cancer, esophageal/gastric, head and neck, testicular/ovarian.
  • Adverse effects include: neuropathy, hearing loss, GI stress, or nausea/vomiting.
  • Nephrotoxicity is the main dose-limiting side effect of Cisplatin, prevented by IV fluids and amifostine (free radical scavenger).
  • Carboplatin has less renal toxicity than Cisplatin.

Topoisomerase Inhibitors

  • Topoisomerase enzymes cut and reseal DNA to relieve tangles and supercoils during replication.
  • Inhibitors lead to DNA breaks without resealing, causing damage and cell death.
  • Affect cells in S and G2 phases (DNA synthesis).
  • Topoisomerase I inhibitors (Irinotecan, Topotecan) are camptothecins from the Camptotheca tree in China.
    • Irinotecan used for colon cancer, Topotecan for ovarian cancer and small cell lung cancer.
  • Adverse effects of Topoisomerase I inhibitors include: myelosuppression and severe diarrhea.
  • Topoisomerase II inhibitors (Etoposide, Teniposide) are synthesized from podophyllotoxins from the May Apple plant.
  • IV and oral use for germ cell cancers, lung cancer (all types), lymphomas.
  • Adverse effects of Topoisomerase II inhibitors include: myelosuppression and nausea/vomiting.

Other Cancer Drugs: Monoclonal Antibodies Overview

  • Monoclonal antibodies are lab produced antibodies from cloned cells in culture
  • Bind to specific antigens (usually surface proteins) on cancer cells for targeted therapy; generally fewer side effects vs traditional chemotherapy.
  • Administered intravenously, often leading to infusion reactions (fever, chills, flushing, itching, skin rashes, GI symptoms) managed with antihistamines or steroids.

Bevacizumab (Avastin)

  • Monoclonal antibody to VEGF-A preventing VEGF-A binding to receptors used in solid tumors like colorectal, breast, or renal cell carcinoma.
  • VEGF (Vascular Endothelial Growth Factor): a signal protein family (VEGF-A/B/C/D); VEGF-A stimulates angiogenesis in tumors.
  • VEGF inhibitors have two scenarios: Bevacizumab for Cancer and Ranibizumab for retinopathy.
  • Toxicity - cardiovascular adverse effects (hypertensions and arterial thromboembolism). Since VEGF System vasodilates via nitric oxide.
  • Other adverse include delayed wound healing and bleeding.

Drugs Interfering With the EGF System

  • EGF (Epidermal Growth Factor) stimulates cell growth and differentiation by binding to EGFR (Epidermal Growth Factor Receptor), a tyrosine kinase receptor.
  • EGF-EGFR binding phosphorylates tyrosine residues, leading to downstream effects for cell growth and differentiation.
  • EGFR is overexpressed in many tumors, making it a therapeutic target.
  • Cetuximab is a monoclonal antibody binding to the EGFR extracellular domain, blocking EGF binding.

Tumors unresponsive to Cetuximab

  • Cetuximab Clinical Study - Tumors even though they expressed EGFR did not respond to cetuximab because of K-ras mutation
  • K-ras is a G-protein downstream EGFR
  • Mutated k-ras has no to benefit from Certuximab, wild-type K-ras respond
  • Result: downstream EGFR with constants activation, no benefit with certuximab

Erlotinib (Tarceva)

  • Oral EGFR tyrosine kinase inhibitor disrupting the EGFR.
  • Benefit in malignancies, mainly non-small cell lung cancer.
  • Side effect: acne-like skin rash (upper torso, face, neck), may indicate drug effect and seen with all EGFR-blocking drugs.

Imatinib

  • Not an antibody, it is a tyrosine kinase inhibitor mainly used in chronic myeloid leukemia (CML).
  • Philadelphia chromosome leads to BCR-ABL fusion gene creating tyrosine kinase protein.
  • Other drugs include dasatinib, nilotinib, and bosutinib.
  • Also combats gastrointestinal stromal tumors (GISTs) associated with c-KIT mutations (treated via surgery with Imatinib)

Skin Rash

  • Skin rash can suggest high Imatinib benefit.
  • GIST tumors are CD 117 positive with gain of function mutation in the kit gene and stem cell factory
  • KIT (also called c-KIT, CD117) is a tyrosine kinase receptor
  • Adverse effects include:
    • Rare pulmonary and peripheral edema
    • Periorbital edema
    • Skin Rash

Rituximab

  • Monoclonal CD20 antibody binding CD20 surface marker on the B-cells; depletes b-cells.
  • Used to treat:
    • Autoimmune diseases
    • Non hodgkin lymphoma
    • CLL (chronic lymphocytic leukemia)
  • ITP
    • First and second line (Steroids, IVIG, spleectomy as an alternative
  • Toxic effect is opportunistic infections Pneumocystis pneumonia, CMV colitis
  • PML = progressive multifocal leukoencephalopathy, caused by the reactivation of JC virus
  • Hepatitis B reactivation with patients

Tamoxifen

  • SERM, selective estrogen receptor modulator is used for estrogen and treatment breast cancer.
  • ER positive is breast cancer treatment.
  • Primary therapy with radiation and chemo.
  • For women who recovered from breast cancer
  • Blocks the estrogen effects in the breast cancer cells, while working as agonist in other tissues.
  • Increase in bone density and endothelium promoting growth
  • Partial agonist to the endothelium , and the effects stimulate polyp, endometrial, and hyperplasia formation
  • Hot flashes is a toxic effect
  • Increase risk is DVT or PE
  • Carcinoma with endometrial associated
  • Raloxifene, also known as Evista can promote an increase in bone density.
  • It works to prevent the osteoporosis in postmenopausal women.

Aromatase Inhibitors

  • Inhibits Anastrozole and Letrozole and EXEmestane
  • Treat and is designed to block stimulation from breast cancer, estrogen receptor as positive
  • Drugs
  • All block estrogen production
  • Side effects
  • Osteoporosis
  • Increase risk of bone fractures from hormone decrease
  • Decreases the amount of the steroids in woman postmenopausal women

Trastuzumab

  • Trade name HERCEPTIN
  • HER 2 is over expressed on the cell receptors, or ERB2, and CD340
  • HER2neu, CD340 HER family HER 2/3/4 receptors as well
  • Breast cancer HER 2
  • By stimulating tumor inhibiting proliferation , tumor dependent
  • Cytotoxicty is antibody
  • Antibody with FC mediated cytotoxicity of tumor dell with system immune, it is important with the cell and killing process
  • No phagoyctosis, no cells , and not toxic as anthracycline chemo
  • Cadiomyopathy and Cardiac effects is associated with the drug
  • It can lead to fall in the ventricular injection factor, with the heart attack factor .

Studying That Suits You

Use AI to generate personalized quizzes and flashcards to suit your learning preferences.

Quiz Team

More Like This

Use Quizgecko on...
Browser
Browser