Download as pdf or txt
Download as pdf or txt
You are on page 1of 126

Anticancer Drugs

Pharmacology Team
Naim Kittana, Suhaib Hattab, Ansam Sawalha, Adham Abu Taha,
Waleed Sweileh, Ramzi Shawahneh

Faculty of Medicine & Health Sciences


An-Najah National University 1
Introduction

• Cancer chemotherapy strives to cause a lethal cytotoxic event or apoptosis


in the cancer cells that can arrest a tumor’s progression.

• The attack is generally directed toward DNA or against metabolic sites


essential to cell replication

• Ideally, these drugs should interfere only with cellular processes that are
unique to malignant cells.

2
Introduction

• Unfortunately, most currently available anticancer drugs do not


specifically recognize neoplastic cells

• Therefore, almost all antitumor agents have a steep dose–response curve


for both therapeutic and toxic effects

3
Principles of cancer chemotherapy
TREATMENT STRATEGIES

4
Goals of treatment

• The ultimate goal of chemotherapy is a cure (that is, long-term, disease-


free survival).

• A true cure requires the eradication of every neoplastic cell.

• If a cure is not attainable, then the goal becomes control of the disease
(stop the cancer from enlarging and spreading) to extend survival and
maintain the best quality of life.

• Thus, the individual maintains a “near-normal” existence, with the cancer


being treated as a chronic disease.

5
Goals of treatment

In either case, the neoplastic cell burden is initially reduced (debulked) by:

• Either surgery and/or by radiation


• Followed by:
 Chemotherapy,
 Immunotherapy: using biological modifiers or
 a combination of these treatment modalities

6
Goals of treatment

• In advanced stages of cancer, the likelihood of controlling the cancer is


not realistic and the goal is palliation (alleviation of symptoms and
avoidance of life-threatening toxicity).

• This means that chemotherapeutic drugs may be used to relieve


symptoms caused by the cancer and improve the quality of life

• In this case the drugs may not extend survival.

• The goal of treatment should always be kept in mind, as it often influences


treatment decisions

7
Goals of treatment

8
Effects of various treatments on the cancer cell burden in a
hypothetical patient

9
Indications for treatment

• Chemotherapy is sometimes used when neoplasms are disseminated and


are not amenable to surgery.

• Adjuvant chemotherapy: when chemotherapy is used as a supplemental


treatment to attack micro-metastases following surgery and radiation

• Neoadjuvant chemotherapy: when chemotherapy is given prior to the


surgical procedure in an attempt to shrink the cancer

• Maintenance chemotherapy: Chemotherapy given in lower doses to assist


in prolonging remission

10
Tumor susceptibility and the growth cycle

• The fraction of tumor cells that are in the


replicative cycle (“growth fraction”)
influences their susceptibility to most
cancer chemotherapeutic agents.

• Rapidly dividing cells are generally more


sensitive to chemotherapy, whereas slowly
proliferating cells are less sensitive

• In general, non-dividing cells (those in the


G0 phase) usually survive the toxic effects
of many of these agents.

11
Tumor susceptibility and the growth cycle

Cell cycle specificity of drugs:


• Both normal cells and tumor cells go through growth cycles.

• However, the number of cells that are in various stages of the cycle may
differ in normal and neoplastic tissues.

• Chemotherapeutic agents that are effective only against replicating cells


are said to be cell cycle specific

• The other agents are said to be cell cycle nonspecific.

• The nonspecific drugs, although having generally more toxicity in cycling


cells, are also useful against tumors that have a low percentage of
replicating cells.

12
Tumor susceptibility and the growth cycle

13
Tumor susceptibility and the growth cycle

Tumor growth rate:

• The growth rate of most solid tumors in vivo is initially rapid, but growth
rate usually decreases as the tumor size increases

• This is due to the unavailability of nutrients and oxygen caused by


inadequate vascularization and lack of blood circulation.

14
Tumor susceptibility and the growth cycle

Tumor growth rate:


Tumor burden can be reduced through:
• Surgery

• Radiation, or
• by using cell cycle–nonspecific drugs to promote the remaining cells into
active proliferation, thus increasing their susceptibility to cell cycle–
specific chemotherapeutic agents.

15
Treatment regimens and scheduling

Log kill phenomenon:


• Destruction of cancer cells by chemotherapeutic agents follows first-order
kinetics (that is, a given dose of drug destroys a constant fraction of cells).

• The term “log kill” is used to describe this phenomenon.

• For example, a diagnosis of leukemia is generally made when there are


about 109 (total) leukemic cells.

• Consequently, if treatment leads to a 99.999% kill, then 0.001% (10-5) of


109 cells (or 104 cells) would remain.

• This is defined as a 5-log kill.


16
Treatment regimens and scheduling

Log kill phenomenon:

• At this point, the patient will become asymptomatic, and the patient is in
remission.

• For most bacterial infections, a 5-log (100,000- fold) reduction in the


number of microorganisms results in a cure, because the immune system
can destroy the remaining bacterial cells.

• However, tumor cells are not as readily eliminated, and additional


treatment is required to totally eradicate the leukemic cell population.

17
Treatment regimens and scheduling

Pharmacologic shelters

• Leukemic or other tumor cells find shelters in tissues such as the CNS,

• Therefore, a patient may require irradiation of the craniospinal axis or


intrathecal administration of drugs.

• Similarly, drugs may be unable to penetrate certain areas of solid tumors

18
Combinations of drugs

• Combination drug chemotherapy is more successful than single-drug


treatment in most of the cancers for which chemotherapy is effective

• Cytotoxic agents in combination are with:


 Qualitatively different toxicities
 Different molecular sites
 Different mechanisms of action,

• Usually combined at full doses

• This results in higher response rates, due to additive and/or potentiated


cytotoxic effects, and nonoverlapping host toxicities

19
Combinations of drugs

• In contrast, agents with similar dose-limiting toxicities, such as


myelosuppression, nephrotoxicity, or cardiotoxicity, can be combined
safely only by reducing the doses of each.

20
Advantages of drug combinations

1) Provide maximal cell killing within the range of tolerated toxicity

2) Effective against a broader range of cell lines in the heterogeneous tumor


population

3) Delay or prevent the development of resistant cell lines.

21
Problems associated with chemotherapy

22
Resistance

• Some neoplastic cells (for example, melanoma) are inherently resistant to


most anticancer drugs.

• Other tumor types may acquire resistance to the cytotoxic effects of a


medication by mutating, particularly after prolonged administration of
suboptimal drug doses.

• The development of drug resistance is minimized by short-term, intensive,


intermittent therapy with combinations of drugs.

• Drug combinations are also effective against a broader range of resistant


cells in the tumor population

23
Multidrug resistance

• P-glycoprotein is responsible for multidrug resistance.

• This resistance is due to ATP– dependent pumping of drugs out of the cell.

• Cross-resistance following the use of structurally unrelated agents also


occurs.

• The presence of P-glycoprotein may account for the intrinsic resistance to


chemotherapy observed with adenocarcinomas.

24
Toxicity

• Therapy aimed at killing rapidly dividing cancer cells also affects normal
cells undergoing rapid proliferation. E.g. cells of :

 The buccal mucosa

 Bone marrow

 GIT mucosa

 Hair follicles

25
Common adverse effects

• Most chemotherapeutic agents have a narrow therapeutic index.


• They can cause:
 Severe vomiting
 Stomatitis
 Bone marrow suppression
 Alopecia

• Vomiting is often controlled by administration of antiemetic drugs.

26
Common adverse effects

• Some adverse reactions are confined to specific agents, such as:


 Bladder toxicity with cyclophosphamide
 Cardiotoxicity with doxorubicin,
 Pulmonary fibrosis with bleomycin.

• The duration of the side effects varies widely.


 Transient: Alopecia
 Irreversible: cardiac, pulmonary, and bladder toxicities

27
Minimizing adverse effects

Some toxic reactions may be ameliorated by interventions, such as:

• The use of cytoprotectant drugs

• Perfusing the tumor locally (for example, a sarcoma of the arm)

• Removing some of the patient’s marrow prior to intensive treatment and


then re-implanting it

28
Minimizing adverse effects

Some toxic reactions may be ameliorated by interventions, such as:

• Promoting intensive diuresis to prevent bladder toxicities.

• The megaloblastic anemia that occurs with methotrexate can be


effectively counteracted by administering folinic acid (leucovorin).

• With the availability of human “granulocyte colony–stimulating factor”


(filgrastim), the neutropenia associated with treatment of cancer by many
drugs can be partially reversed.

29
Problems associated with chemotherapy

Treatment-induced tumors:
• Because most antineoplastic agents are mutagens, neoplasms (for
example, acute nonlymphocytic leukemia) may arise 10 or more years
after the original cancer was cured.

• Treatment-induced neoplasms are especially a problem after therapy with


alkylating agents

• Most tumors that develop from cancer chemotherapeutic agents respond


well to treatment strategies

30
Anticancer drugs

ANTIMETABOLITES

31
ANTIMETABOLITES

• Antimetabolites are structurally related to normal compounds that exist


within the cell.

• They generally interfere with the availability of normal purine or


pyrimidine nucleotide precursors, either by:
 Inhibiting their synthesis or
 Competing with them in DNA or RNA synthesis.

• Their maximal cytotoxic effects are in S-phase and are, therefore, cell cycle
specific.

32
Methotrexate (MTX)

• MTX is structurally related to folic acid and acts


as an antagonist of the vitamin by inhibiting
mammalian dihydrofolate reductase.

• The inhibition of DHFR can only be reversed by:


 a 1000-fold excess of the natural substrate,
dihydrofolate (FH2), or

 by administration of leucovorin, which


bypasses the blocked enzyme and replenishes
the folate pool

33
ANTIMETABOLITES

• Leucovorin, or folinic acid, is the N5-formyl group–carrying form of FH4.

• MTX is specific for the S-phase of the cell cycle.

• Pemetrexed is an antimetabolite similar in mechanism to methotrexate.

• However, in addition to inhibiting DHFR, it also inhibits thymidylate


synthase and other enzymes involved in folate metabolism and DNA
synthesis.

• Pralatrexate is a newer antimetabolite that also inhibits DHFR

34
Therapeutic uses of MTX

MTX, usually in combination with other drugs, is effective against:


• Acute lymphocytic leukemia (ALL)

• Burkitt lymphoma in children

• Breast cancer

• Bladder cancer

• Head and neck carcinomas.

35
Therapeutic uses of MTX

Low-dose MTX is effective as a single agent against certain inflammatory


diseases as:
• Severe psoriasis

• Rheumatoid arthritis

• Crohn disease.

36
Pharmacokinetics of MTX

• MTX is administered orally, IM, IV, and intrathecally (does not penetrate
BBB).

• Distributes well to intestinal epithelium, liver, kidney, skin as well as in


ascites, pleural effusions.

• High doses results in crystalluria; avoid renal toxicity by keeping the urine
alkaline and the patient well hydrated.

37
Side effects of MTX

• N/V/D
• Stomatitis
• Rash
• Alopecia
• Myelosuppression
• High-dose: renal damage
• Intrathecal: neurologic toxicities

38
6-Mercaptopurine (6-MP)

• Thiol analog of hypoxanthine purine analog

Clinical use:
• Maintenance of remission in acute lymphoblastic leukemia.

• 6-MP and its analog, Azathioprine, can be used for Crohn disease

39
Mechanism of action of 6-MP

1. Nucleotide formation:
6-MP must penetrate target cells and be converted to the nucleotide
analog, 6-MP-ribose phosphate (TIMP).

2. Inhibition of purine synthesis: TIMP also blocks the formation of


adenosine monophosphate

40
Mechanism of action of 6-MP

3. Incorporation into nucleic acids:


 TIMP is converted to thioguanine monophosphate, which after
phosphorylation to di- and triphosphates can be incorporated into RNA.

 The deoxyribonucleotide analogs that are also formed are incorporated


into DNA.

 This results in nonfunctional RNA and DNA

41
Fludarabine

• Is a purine nucleotide analog.

• It is useful in the treatment of:


 Chronic lymphocytic leukemia (CLL)

 Hairy cell leukemia

 Indolent non-Hodgkin lymphoma

42
Mechanism of action Fludarabine

• Fludarabine is a prodrug,

• It is phosphorylated into triphosphate that is incorporated into both DNA


and RNA, decreases their synthesis in the S-phase

43
Cladribine

Mechansim of action of Cladribine


• Purine analog

• Undergoes reactions similar to those of Fludarabine

• It must be phosphorylated to a nucleotide to be cytotoxic.

• It becomes incorporated at the 3′-terminus of DNA and, thus, hinders


elongation.

• It also affects DNA repair and is a potent inhibitor of ribonucleotide


reductase.

44
Cladribine

• Spectrum of activity: similar to those of Fludarabine.

• Cladribine distributes throughout the body, including into the CSF.

45
5-Fluorouracil (5-FU)

Clinical use of 5-FU: treatment of slowly growing


solid tumors, e.g.:
• Colorectal
• Breast
• Ovarian
• Pancreatic
• Gastric
• Topically, effective for the treatment of
superficial basal cell carcinomas.

5-FU = 5-fluorouracil
5-FUR = 5-fluorouridine
5-FUMP = 5-fluorouridine monophosphate
5-FUDP = 5-fluorouridine diphosphate
5-FUTP = 5-fluorouridine triphosphate
dUMP = deoxyuridine monophosphate
dTMP = deoxythymidine monophosphate 46
Cytarabine

Mechanism of action:

• Acts as a pyrimidine antagonist.

• Must be phosphorylated to the nucleotide form (cytosine arabinoside


triphosphate or ara-CTP) to be cytotoxic.

• Ara-CTP is an effective inhibitor of DNA polymerase.

• The nucleotide is also incorporated into nuclear DNA and can terminate
chain elongation.

The major clinical use : in acute nonlymphocytic (myelogenous) leukemia (AML).

47
Gemcitabine

• It is used most commonly for:


– Pancreatic cancer
– Non– small cell lung cancer

48
Anticancer drugs

ANTIBIOTICS

49
Antibiotics

Their cytotoxic action is primarily due to:


• Their interactions with DNA, leading to disruption of DNA function.
• Their abilities to inhibit topoisomerases (I and II)
• Their ability to produce free radicals.

• All are cell cycle nonspecific except Bleomycin.

50
Anthracyclines

Common examples
• Doxorubicin
• Daunorubicin
• Idarubicin
• Epirubicin
• Mitoxantrone

51
Clinical uses of Anthracyclines

• Doxorubicin is used in combination with other agents for treatment of:


 Breast and lung cancers
 Acute lymphocytic leukemia and lymphomas.

• Daunorubicin and idarubicin: are used for Acute leukemias

• Mitoxantrone: is used in prostate cancer.

52
Mechanism of action of Anthracyclines

Produce free radicals that induce


• Membrane lipid peroxidation

• DNA strand scission

• Direct oxidation of purine/ pyrimidine


bases, thiols, and amines

53
Adverse effects of Anthracyclines

• Cardiotoxicity
 Irreversible and dose-dependent
 Caused by free radicals and lipid peroxidation (the most serious
adverse reaction)
 More common with Daunorubicin and Doxorubicin than with
Idarubicin and Epirubicin.

• Extravasation is a serious problem that can lead to tissue necrosis

54
Bleomycin

• Cause scission of DNA by an oxidative


process.

• It is cell cycle specific and causes cells


to accumulate in the G2 phase.

• Primary clinical use: Treatment of


testicular cancers and Hodgkin
lymphoma

55
Adverse effects of Bleomycin

• Pulmonary toxicity is the most serious adverse effect, progressing from


rales, cough, and infiltrate to potentially fatal fibrosis

• Mucocutaneous reactions (common)

• Alopecia (common)

56
Anticancer drugs

ALKYLATING AGENTS

57
Alkylating agents

• Covalently bind to nucleophilic groups on various cell constituents.

• Alkylation of DNA is the crucial cytotoxic reaction that is lethal to the


tumor cells.

• Alkylating agents do not discriminate between cycling and resting cells,

• They are most toxic for rapidly dividing cells.

58
Alkylating agents

• They are used in combination with other agents to treat a wide variety of
lymphatic and solid cancers.

• All are mutagenic and carcinogenic and can lead to secondary


malignancies such as acute leukemia

59
Cyclophosphamide and Ifosfamide

• Used either singly or as part of a regimen


in the treatment of a wide variety of
neoplastic diseases, such as
 Non-Hodgkin lymphoma
 Sarcoma
 Breast cancer

• Cyclophosphamide can be given


orally or IV

• Ifosfamide is IV only

60
Adverse effects of Cyclophosphamide and Ifosfamide

• Hemorrhagic cystitis
 Can lead to fibrosis of the bladder.

 Due to the excretion of the toxic metabolite acrolein in the urine.

 Reduced by:
– Adequate hydration and
– IV injection of mesna (neutralizes acrolein)

• High incidence of neurotoxicity

61
Nitrosoureas

• Carmustine and Lomustine

• Can penetrate CNS, thus primarily employed in the treatment of brain


tumors

• More cytotoxic to dividing cells

62
Other alkylating agents

• Chlorambucil, Melphalan and Busulfan

• Used in lymphatic cancers

• Have moderate hematologic toxicities and upset the GI tract

• Busulfan can cause pulmonary fibrosis

• All of these agents are leukemogenic

63
Anticancer drugs
MICROTUBULE INHIBITORS

64
Microtubules inhibitors

• The mitotic spindle is essential for the movements of structures occurring


in the cytoplasm of all eukaryotic cells.

• The mitotic spindle consists of chromatin plus a system of microtubules


composed of tubulin.

• The mitotic spindle is essential for the equal partitioning of DNA into the
two daughter cells that are formed when a eukaryotic cell divides.

• These drugs disrupt this process by affecting the equilibrium between the
polymerized and depolymerized forms of the tubulin, thereby causing
cytotoxicity

65
Vincristine (VX) and Vinblastine (VBL)

• Derived Vinca rosea plant (Vinca alkaloids)

• Structurally similar to one another, but their therapeutic indications are


different

66
Mechanism of action of Vinca alkaloids

They are cell


cycle specific
and phase
specific

67
Clinical uses of Vincristine (VX)

• Acute lymphoblastic leukemia in children

• Wilms tumor

• Ewing soft tissue sarcoma

• Hodgkin and non-Hodgkin lymphomas

• Some other rapidly proliferating neoplasms

68
Clinical uses of Vinblastine (VBL)

• Metastatic testicular carcinoma: Administered with Bleomycin and


Cisplatin

• Systemic Hodgkin and non-Hodgkin lymphomas.

• Advanced non–small cell lung cancer: either as a single agent or with


Cisplatin

69
Adverse effects of Vinca alkaloids

• Both VX and VBL cause:


 Phlebitis or cellulitis, if the drugs extravasate during injection
 Nausea, vomiting & diarrhea
 Alopecia

• VBL is a more potent myelosuppressant than VX

• Peripheral neuropathy (paresthesias, loss of reflexes, foot drop, and


ataxia) is associated with VX

• Can be fatal if administered intrathecally

70
Paclitaxel and docetaxel

Microtubules stabilizers

71
Adverse effects of Paclitaxel and docetaxel

• Neutropenia and leukopenia.

• Alopecia

• Vomiting and Diarrhea (uncommon)

• Serious hypersensitivity reactions: Should premedicate patient with


Dexamethazone and Diphenhydramine

72
Anticancer drugs
STEROID HORMONES AND THEIR ANTAGONISTS

73
Strategies for the treatment of steroid-sensitive tumors

Tumors that are steroid hormone sensitive may be either


1) Hormone responsive: the tumor regresses following treatment with a
specific hormone or

2) Hormone dependent: Removal of a hormonal stimulus causes tumor


regression; or

3) Both

74
Strategies for the treatment of steroid-sensitive tumors

Removal of hormonal stimuli from hormone-dependent tumors can be


accomplished :

• Surgically: e.g. orchiectomy or castration) or

• Pharmacologically: e.g. antiestrogens

75
Prednisone

• Potent, synthetic, anti-inflammatory corticosteroid with less


mineralocorticoid activity than cortisol

• At high doses, cortisol is lymphocytolytic and leads to hyperuricemia due


to the breakdown of lymphocytes

• Prednisone is a prodrug that is bioactivated into Prednisolone by the liver

76
Prednisone

Prednisone is primarily employed to induce remission of:


• Acute lymphocytic leukemia (ALL)
• Hodgkin lymphomas
• non-Hodgkin lymphomas

77
Tamoxifen

• It is classified as a selective estrogen receptor modulator (SERM)

• It is an estrogen antagonist with some estrogenic activity

• first-line therapy in the treatment of estrogen receptor–positive breast


cancer

• Used prophylactically in reducing breast cancer occurrence in women who


are at high risk.

• Can stimulate premalignant lesions due to its estrogenic properties:


patients should be closely monitored during therapy.

78
Mechanism of action of Tamoxifen

79
Mechanism of action of Tamoxifen

• Binds to estrogen receptors in the breast tissue, but the complex is unable
to translocate into the nucleus for its action

• The result is a depletion (down-regulation) of estrogen receptors

• Estrogen competes with Tamoxifen. Therefore, in premenopausal women,


the drug is used with a gonadotropin releasing hormone (GnRH) analog
such as Leuprolide, which lowers estrogen levels.

80
Adverse effects of Tamoxifen

• Hot flashes

• Nausea & vomiting

• Vaginal bleeding & discharge (due to estrogenic activity of the drug and
some of its metabolites).

• Hypercalcemia

• Increased pain if the tumor has metastasized to bone

• Thromboembolism

• May cause endometrial cancer

81
Raloxifene

• It is a SERM given orally that acts to block estrogen effects in the uterine
and breast tissues, while promoting effects in the bone to inhibit
resorption.

• Reduce the risk of estrogen receptor– positive invasive breast cancer in


postmenopausal women

82
Fulvestrant

• An estrogen receptor antagonist

• Given to patients with hormone receptor– positive metastatic breast


cancer.

• Binds to and causes estrogen receptor down-regulation on tumors and


other targets

83
Aromatase inhibitors

• Anastrozole and Letrozole

• The aromatase reaction is responsible for the extra-adrenal synthesis of


estrogen, which takes place in:
 liver, fat, muscle, skin, and breast tissues, including breast
malignancies

• Peripheral aromatization is an important source of estrogen in


postmenopausal women.

• Aromatase inhibitors decrease the production of estrogen in these women

84
Anastrozole and letrozole

• Nonsteroidal aromatase inhibitors.

• They do not predispose patients to endometrial cancer

• They are devoid of the androgenic side effects

• in the United States they are considered second-line therapy after


Tamoxifen for hormone dependent breast cancer

• They have become first-line drugs in other countries for the treatment of
breast cancer in postmenopausal women.

• They are orally active and cause almost a total suppression of estrogen
synthesis

85
Aromatase inhibitors & Tamoxifen for Breast cancer therapy

86
Progestins

• Megestrol used in treating metastatic hormone-responsive breast and


endometrial neoplasms

• Aromatase inhibitors are replacing it in therapy

87
Leuprolide, Goserelin, and Triptorelin

• Synthetic analogs of GnRH

• Continuous administration leads to receptor desensitization and,


consequently, inhibition of FSH and LH release

• LH stimulus for the secretion of testosterone by the testes

• FSH stimulates the secretion of estrogen

• These drugs reduce both androgen and estrogen syntheses

88
Leuprolide, Goserelin, and Triptorelin

• Response to Leuprolide in prostatic cancer is equivalent to that of


orchiectomy with regression of tumor and relief of bone pain

• They are beneficial for women with advanced breast cancer and have
largely replaced estrogens in therapy for prostate cancer

• They are available as:


 Sustained-release intradermal implant
 Subcutaneous depot injection
 Intramuscular depot injection

89
Adverse effects of GnRH analogues

• Impotence

• Hot flashes

• Tumor flare

90
Estrogens

• Example: Ethinyl estradiol

• Used in the treatment of prostatic cancer.

• They have been largely replaced by the GnRH analogs because of fewer
adverse effects.

• Estrogens inhibit the growth of prostatic tissue by blocking the production


of LH, thereby decreasing the synthesis of androgens in the testis

91
Side effects of Estrogens

• Thromboemboli

• Myocardial infarction

• Strokes

• Hypercalcemia

• Men who are taking estrogens may experience gynecomastia and

impotence

92
Nonsteroidal antiandrogens

• Flutamide, Nilutamide, and Bicalutamide

• They compete with the natural hormone for binding to the androgen
receptor and prevent its translocation into the nucleus

• Used in the treatment of prostate cancer

• Side effects:
 Gynecomastia
 GI distress
 Liver failure (Rare)

93
Anticancer drugs
MONOCLONAL ANTIBODIES

94
Trastuzumab

• Used for metastatic breast cancer, overexpressing transmembrane


“human epidermal growth factor receptor protein 2” (HER2)

• HER2 is overexpressed in 25% to 30% of patients with breast cancer

• HER2 can be also expressed in gastric and gastroesophageal cancers

95
Mechanism of action of Trastuzumab

• Binds to HER2 receptors and inhibits cell proliferation

• It blocks downstream signaling pathways

• It induces antibody-dependent cytotoxicity

96
Adverse effects of Trastuzumab

• Congestive heart failure

• Cardiotoxicity is worsened if given in combination with anthracyclines

97
Rituximab

• Directed against the CD20 antigen that is found on the surfaces of normal
and malignant B lymphocytes

• CD20 antigen is expressed on nearly all B-cell non-Hodgkin lymphomas but


not in other bone marrow cells

• CD20 plays a role in the activation process for cell cycle initiation and
differentiation

98
Mechanism of action of Rituximab

• Upon binding to CD20, the Fc domain of the antibody induces


complement and antibody-dependent, cell-mediated cytotoxicity against B
cells

• Rituximab is effective in the treatment of:


 Lymphomas
 Chronic lymphocytic leukemia
 Rheumatoid arthritis

99
Adverse effects of Rituximab

• Severe fatal hypersensitivity reactions, especially in patients with high


circulating levels of neoplastic cells

• Occurs due to rapid activation of complement which results in the release


of tumor necrosis factor-α (TNF-α) and interleukins

• Pretreatment with diphenhydramine, acetaminophen, and corticosteroids


can ameliorate these problems

100
Adverse effects of Rituximab

• Tumor lysis syndrome: Occurs within 24 hours of the first dose of


Rituximab. Consists of:

 Hyperkalemia
 Hypocalcemia
 Hyperuricemia
 Hyperphosphatasemia
 Acute renal failure that may require dialysis.

101
Bevacizumab

• An IV antiangiogenesis agent.

• Directed against vascular endothelial growth factor (VEGF)

• Stops VEGF from stimulating the formation of new blood vessels


(neovascularization).

• Without new blood vessels, tumors do not receive the oxygen and
essential nutrients necessary for growth and proliferation.

102
Clinical uses of Bevacizumab

• Approved for use as a first-line drug against metastatic colorectal cancer in


combination with 5-FU–based chemotherapy.

• Diabetic retinopathy (intravitreous injection)

103
Cetuximab

• Targets the epidermal growth factor receptor (EGFR) on the surface of


cancer cells and interfering with their growth

• Approved to treat KRAS wild-type metastatic colorectal cancer and head


and neck cancers

104
Anticancer drugs
PLATINUM COORDINATION COMPLEXES

105
Drugs

• Cisplatin

• Carboplatin

• Oxaliplatin

106
Mechanism of action

• Bind to guanine in DNA, forming inter- and intrastrand crosslinks.

• The resulting cytotoxic lesion inhibits both polymerases for DNA


replication and RNA synthesis.

• Cytotoxicity can occur at any stage of the cell cycle, but cells are most
vulnerable to the actions of these drugs in the G1 and S-phases.

107
Clinical use

• Metastatic testicular carcinoma in combination with VBL and bleomycin

• Ovarian carcinoma in combination with cyclophosphamide

• Bladder carcinoma: alone

• Oxaliplatin is used in the setting of colorectal cancer.

108
Adverse effects

• Severe, persistent vomiting occurs for at least 1 hour after administration


of Cisplatin and may continue for as long as 5 days.
 Premedication with antiemetic agents is required.

• The major limiting toxicity of Cisplatin is dose-related nephrotoxicity


 Can be prevented by aggressive hydration.

• Ototoxicity with high-frequency hearing loss and tinnitus.

109
Adverse effects

• Unlike Cisplatin, Carboplatin causes only mild nausea and vomiting, and it
is rarely nephro-, neuro-, or ototoxic

• Myelosuppression

• Hepatotoxicity

• Anaphylaxis

110
Anticancer drugs
TOPOISOMERASE INHIBITORS

111
Topoisomerase inhibitors

• They inhibit topoisomerase enzymes that reduce supercoiling of DNA

Drugs

• Camptothecins
 Irinotecan and Topotecan
 They are plant semisynthetic alkaloids of Camptotheca tree

• Etoposide
 Semisynthetic derivative of the plant alkaloid, podophyllotoxin

112
Mechanism of action of Camptothecins

113
Camptothecins

• Topotecan is used in
 metastatic ovarian cancer when primary therapy has failed
 treatment of small cell lung cancer.

• Irinotecan is used with 5-FU and leucovorin for the treatment of colorectal
carcinoma.

114
Etoposide

Mechanism of action

• The drug binds the enzyme-DNA complex

• This results in persistence of the transient cleavable form of the complex


and causes irreversible double-strand breaks.

115
Mechanism of action of Etoposide

116
Etoposide

• Clinical use:
 Treatment of lung cancer
 Testicular carcinoma: in combination with bleomycin and cisplatin.

• Major toxicity: Dose-limiting myelosuppression (primarily leukopenia)

117
Anticancer drugs
TYROSINE KINASE INHIBITORS

118
Imatinib

• A deregulated BCR-ABL kinase is present in the leukemia cells of almost


every patient with CML and GI stromal tumors

• BCR-ABL kinase is associated with cell proliferation

• Imatinib is designed to inhibit this mutated enzyme

119
Erlotinib

• an inhibitor of the Epidermal Growth Factor Receptor Tyrosine Kinase.

• Approved for the treatment of non–small cell lung cancer and pancreatic
cancer

120
Anticancer drugs
MISCELLANEOUS AGENTS

121
Procarbazine

• Inhibits DNA, RNA, and protein synthesis


• Distributes rapidly to the CNS

Side effects:
• Bone marrow depression (major toxicity)

• Nausea, vomiting, and diarrhea (common)

• Neurotoxicity: drowsiness, hallucinations and paresthesias


• Mutagenic and teratogenic: Nonlymphocytic leukemia has developed in
patients treated with the drug.
• It inhibits MAO: avoid with foods containing tyramine

122
Asparaginase

• Some neoplastic cells require an external source of asparagine because of


limited capacity to synthesize sufficient amounts of the amino acid to
support growth and function

• Asparaginase catalyzes the deamination of asparagine to aspartic acid and


ammonia, thus depriving the tumor cells of this amino acid, which is
needed for protein synthesis

• Clinical use: treatment of childhood acute lymphocytic leukemia in


combination with VX and prednisone

123
Interferons

• α-interferons: produced by leukocytes

• β- interferons: produced by connective tissue fibroblasts

• γ- interferons: produced by T lymphocytes

• interferon-α-2a and 2b: are produced by Recombinant DNA techniques

124
Interferons

• Interferon-α-2a is approved for the management of hairy cell leukemia,


CML, and AIDS-related Kaposi sarcoma

• Interferon-α-2b is approved for the treatment of hairy cell leukemia,


melanoma, AIDS-related Kaposi sarcoma, and follicular lymphoma

• the exact mechanism by which the interferons are cytotoxic is unknown.

125
126

You might also like