Cancer is characterized by the transformation of normal cells to ones characterized by abnormal cellular differentiation, proliferation, invasion, and metastases. The molecular and biochemical bases underlying the transformation process are becoming increasingly clear and provide critically important information for identifying new drug targets.

Normal cell division results from the interaction of growth factors with specific receptors (plasma membrane, cytoplasmic, nuclear). This initiates a signal transduction cascade through receptor tyrosine kinases and downstream serine, threonine kinases that culminates in uncoiling of DNA by the action of histone acetylases and topoisomerases and activation of nuclear transcription factors that produce cell-proliferation and cell-viability molecules. It should not, therefore, be surprising that cancer cells usurp these normal pathways and that our most effective drugs target many of these processes ( Figure 46-1 ).

Figure 46-1
Targets for anticancer drugs
Anticancer drugs work by interfering with the processes underlying normal cellular physiology. These include receptor-activated signal transduction pathways culminating in transcriptional activation, DNA replication, protein synthesis, and cell division.

(Modified by permission from Macmillan Publishers Ltd: Downward J. The ins and outs of signaling. Nature . 2001;411:759).

Malignant cells acquire the ability to replicate indefinitely, invade, and metastasize. This process includes activation of telomerase, detachment from the primary site, anchorage-independent growth, invasion through the basement membrane, and access to the blood or lymphatic vessels, as well as entry to distant organs through adherence to visceral capillaries and the ability to grow in a foreign site, thereby escaping a variety of immunological mechanisms designed to protect the host from “foreign invasion.” An understanding of the interactions between cancer cells and the surrounding stroma (malignant tissue) helped identify new targets to interfere with this characteristic of malignancy.

Recent attention has been turned to the existence and role of cancer stem cells. Dick and colleagues first identified cells from acute myeloid leukemia that had stem-cell characteristics based on the identification of a subpopulation of cells with a CD34 + /CD38 phenotype that possessed the ability to recapitulate the phenotypic heterogeneity of the original leukemia. Subsequently, cancer stem cells (CSCs) or tumor-initiating cells have been found in other leukemias and in most solid tumors. The therapeutic importance of the stem cell model is that it posits that our inability to cure most tumors is due to resistance of CSCs to current chemotherapy and radiation therapy.

In this chapter, we attempt to place cancer chemotherapeutic drugs in a molecular biological context as summarized in Tables 46-1, 46-2, and 46-3 .

We focus primarily on drugs that have achieved approval from the U.S. Food and Drug Administration (FDA); numerous drugs against new targets are in development, but a comprehensive description is beyond the scope of this chapter.

Table 46-1
Chemotherapeutic Drug Overview
Target Examples Use
Nuclear Receptors
Estrogen receptor Tamoxifen, toremifene, raloxifene, fulvestrant Treatment and prevention of breast cancer
Progesterone receptor Megestrol acetate Breast cancer
Retinoid receptor Retinoic acid Promyelocytic leukemia
Androgen receptor Bicalutamide, flutamide, nilutamide, enzalutamide Prostate cancer
Plasma Membrane Receptors and Tyrosine Kinases
Her-1 (EGFR) Gefitinib, erlotinib, cetuximab, panitumumab Non–small-cell lung; pancreas
Her-2/neu Trastuzumab, pertuzumab Breast
Bcr:abl Imatinib, dasatinib, nilotinib Chronic myelogenous leukemia
VEGF receptor Sorafenib, sunitinib Renal cell
cKit Imatinib, sorafenib, sunitinib Gastrointestinal stromal tumor (GIST)
Flt-3 Sorafenib, sunitinib
Fms Sunitinib
Gonadotropin receptors Abarelix, leuprolide, goserelin
Growth factors
Aromatase inhibitors
Cyp17A1
Letrozole, anastrozole, exemestane
Abiraterone acetate
Breast cancer
Prostate cancer
Vascular endothelial growth factor (VEGF) Bevacizumab Colorectal cancer
Multiple Kinases
VEGFR, PDGFR, cKIT, Flt-3, FMS, bRAF, Ret Sunitinib Renal; GIST
VEGFR, PDGFR, cRAF, bRAF, Flt-3, Fms Sorafenib Renal
Miscellaneous
CD20
CD25
Rituximab, ibritumomab tiuxetan (Zevalin), tositumomab (Bexxar), ofatumumab
Denileukin diftitox
Lymphoid malignancies
Cutaneous T-cell lymphoma
CD52
CD30
Alemtuzumab
Brentuximab vedotin
Chronic lymphocytic leukemia (CLL)
Hodgkin’s disease
DNA Synthesis
Dihydrofolate reductase Methotrexate, trimetrexate Breast, lymphocytic leukemia, choriocarcinoma, lymphoma
Thymidylate synthase 5-Fluorouracil, capecitabine, pemetrexed Colon, breast, mesothelioma
Adenosine deaminase Pentostatin, cladribine (2CDA) Hairy-cell leukemia, lymphomas, CLL
DNA Replication
Nucleic acid bases
Alkylating agents Nitrogen mustards (e.g., mechlorethamine, cyclophosphamide, bendamustine) Leukemia, lymphoma, breast, brain, melanoma, etc.
Nitrosoureas (e.g., BCNU, temozolomide) Glioblastoma, anaplastic astrocytoma
Ethyleneimines (e.g., thiotepa) Bone marrow transplantation
Alkyl sulfonates (e.g., busulfan) Leukemia
Triazenes (e.g., dacarbazine) Melanoma
Platinating agents Cis-, carbo-, oxaliplatin Lung, head and neck, bladder, germ cell, colorectal
Transcription inhibitors Actinomycin-D Wilms’ tumor
DNA methylation 5′-Azacytidine, decitabine Myelodysplastic syndrome
Topoisomerases
Topoisomerase I Topotecan, irinotecan Colorectal, ovary, lung, cervical
Topoisomerase II Doxorubicin, epirubicin, etoposide, mitoxantrone Breast, lymphoma, leukemia, lung, ovary, testicular, etc.
Microtubules
Vinca alkaloids Vincristine, vinblastine, vinorelbine Breast, lung, acute lymphocytic leukemia (ALL), bladder, lymphoma, etc.
Taxanes
Epothilones
Paclitaxel, docetaxel
Ixabepilone
Breast, lung, bladder, ovarian, etc.
Breast cancer
Protein Synthesis
l -Asparaginase Childhood ALL, T-cell lymphoma
Protein Degradation
60S proteasome Bortezomib, carfilzomib Multiple myeloma, mantle-cell lymphoma

Table 46-2
Drugs That Alter Nucleic Acid Synthesis and Function
Target Examples Use
DNA Synthesis
Dihydrofolate reductase Methotrexate, trimetrexate, pemetrexed, pralatrexate Breast, lymphocytic leukemia, choriocarcinoma, lymphoma, etc.
Thymidylate synthase 5-Fluorouracil, capecitabine, pemetrexed Colon, breast, mesothelioma, etc.
Adenosine deaminase Pentostatin, cladribine (2CDA) Hairy-cell leukemia, lymphomas, CLL
DNA Replication
Nucleic acid bases
Alkylating agents Nitrogen mustards (e.g., mechlorethamine, cyclophosphamide) Leukemia, lymphoma, breast, brain, melanoma, etc.
Nitrosoureas (e.g., BCNU)
Ethyleneimines (e.g., thiotepa)
Alkyl sulfonates (e.g., Busulfan)
Triazenes (e.g., dacarbazine, temozolomide)
Platinating agents Cis, carbo-, oxaliplatin Lung, head and neck, bladder, germ cell, colorectal
Transcription inhibitors Actinomycin-D Wilms’ tumor
DNA methylation 5′-Azacytidine Myelodysplastic syndrome
Topoisomerases
Topoisomerase 1 Topotecan, irinotecan Colorectal, ovary, lung, cervical
Topoisomerase II Doxorubicin, epirubicin, etoposide, mitoxantrone Lymphoma, leukemia, lung, ovary, testicular

Table 46-3
Drugs with Other Mechanisms
Target Examples Use
Microtubules
Vinca alkaloids Vincristine, vinblastine, vinorelbine Breast, lung, ALL, bladder, lymphoma
Taxanes Paclitaxel, docetaxel Breast, lung, bladder, ovarian
Protein Synthesis
l -Asparaginase Childhood ALL, T-cell lymphoma
Protein Degradation
60S proteasome Bortezomib Multiple myeloma, mantle-cell lymphoma

Molecular Basis of the Therapeutic Index

Therapeutic index is defined as follows: LD 50 , or median lethal dose, is the dose of drug that causes death in 50% of experimental animals, and ED 50 , or median effective dose, is the dose that produces a specified effect (“response”) in 50% of the population under study. The therapeutic index in the clinic compares the dose of a drug that causes untoward toxicities to the dose that produces the desired therapeutic effect.

All drugs have targets, but it is the unique relationship of the target to the disease that can ultimately affect a drug’s therapeutic index. Traditional drug targets included DNA (nucleotide bases, enzymes of DNA synthesis, degradation, and repair), microtubules, and growth factor receptors. New targets include mutated, overexpressed, or fused growth factor/oncogene products (EGFR [Her-1], Her-2/neu, ras, bRaf, bcr:abl), immune checkpoint modulators (CTLA4, PD-1), cell surface antigens (CD33, CD22, CD20), anti-apoptotic proteins (bcl-2), cell-cycle regulators (cyclin-dependent kinases), epigenetic targets (histone deacetylases and methyltransferases), metabolic pathways (mTOR, PI3K, AKT) , stem-cell pathways (notch, wnt), and the machinery of protein synthesis ( l -asparaginase) and degradation (proteasome). Drugs that affect these newer targets are often referred to as targeted therapies , creating the false impression that classic chemotherapeutics do not have targets.

The specificity/selectivity of a drug for a particular target is, in general, proportional to the affinity constant, K a , more accurately defined for competitive drug target interactions as the K i , which is the reciprocal of the concentration of drug required to inhibit 50% of the target’s activity when controlled for all possible ligand or substrate concentrations. The activity of a drug refers to its effectiveness independent of dose or concentration.

There are several factors that help explain why cancer cells are more sensitive to cancer therapeutic drugs than normal tissues. For any drug, the therapeutic index is related to absorption, uptake, distribution, and metabolism. Differences in tumor vasculature, intratumoral pressure, and drug binding may alter drug uptake in a favorable or unfavorable way. Classically, the therapeutic index of intravenously administered cancer chemotherapy has been thought to be due primarily to cell-cycle kinetics. Many chemotherapeutic agents are more effective against cycling than noncycling cells and are tested under cell culture conditions where cancer cells rapidly proliferate (“log phase”); this a posteriori conclusion was derived from these observations. However, many solid tumors have a relatively long doubling time, yet a therapeutic index remains. Therefore, alternative explanations must exist. One includes differences in energy requirements between normal and malignant cells. For example, whereas normal tissues use oxidative phosphorylation to metabolize glucose, malignant tissues are often dependent on aerobic glycolysis. This is thought to reflect the selection pressure placed on tumor cells to cope with relatively hypoxic and nutrient-deprived conditions. Rather than using the electron transfer chain within the mitochondria to yield 36 mol ATP per mol glucose, cancer cells metabolize glucose via glycolysis, generating a net 2 mol ATP per mol glucose metabolized (the Warburg effect). As a result, cancer cells are metabolically fragile and are unable to cope as readily with cellular damage. This may be particularly relevant for drugs that block the effects of growth factors, because growth factor depletion produces rapid downregulation of nutrient transporters, which would lead to metabolic crisis in cancer cells more rapidly than in normal cellular counterparts.

Another concept is that tumor cells exhibit “oncogene addiction” and that inhibition of one or more of these oncogene products rapidly results in apoptotic cell death in “addicted” cancer cells but not in normal counterparts. Thus, tumor cells that depend on their survival by overexpression of a growth factor receptor (e.g., EGFR) would be more susceptible to inhibitors than are normal cells.

Drugs Affecting Growth Factors and Growth Factor Receptors

Cancer cells usurp the mechanisms of normal cell division, which results from the interaction of growth factors with specific receptors (plasma membrane, cytoplasmic, or nuclear). This initiates a signal transduction cascade culminating in activation of nuclear transcription factors that produce cell-proliferation and cell-viability molecules. Thus, it stands to reason that some of our most effective drugs target growth factors or their receptors (see Table 46-1 ) and the downstream consequences of this interaction that include activation of protein kinases, replication of DNA, transcription of mRNA, synthesis of new proteins, formation of the mitotic spindle through microtubule polymerization, and creation of interphase daughter cells via microtubule depolymerization (see Table 46-2 ). In addition, the nutrient requirements of cancer cells create an increased dependence on the uptake of glucose (the basis for positron emission tomography or PET scan) and the ability to sustain energy requirements through frequent alterations in the PI3K/AKT/mTOR pathway and activation of autophagic cell survival ( Figure 46-2 ).

Figure 46-2, Energy regulation through PI3 kinase signaling

Drugs Affecting Growth Factors

Sex Hormones

Aromatase is an enzyme complex made up of two proteins, aromatase cytochrome P450 (CYP19) and NADPH-cytochrome P450 reductase. Inhibition of aromatase blocks the conversion of androgens (androstenedione) to estrone in peripheral tissues including fat, liver, muscle, and breast without detectable effects on adrenal synthesis of corticosteroids or aldosterone. Following the reports by Santen and colleagues that aminoglutethimide could inhibit the conversion of androstenedione to estradiol, aromatase became an attractive target for new drug development. Three aromatase inhibitors are used in the clinic, including letrozole (Femara), anastrozole (Arimidex), and exemestane (Aromasin). Whereas letrozole and anastrozole are reversible, nonsteroidal inhibitors of aromatase, exemestane is a steroidal derivative of androstenedione that binds irreversibly to the enzyme and targets the protein for degradation. Aromatase inhibitors further deplete circulating estradiol in postmenopausal women and are highly effective in the treatment of breast cancer in the adjuvant and metastatic settings. These well-tolerated medications can produce osteopenia and are often prescribed with a bisphosphonate, calcium, and vitamin D to prevent this complication.

Cyp17A1 is a cytochrome P450 enzyme complex that has both hydroxylase and lyase activity. It catalyzes the hydroxylation of pregnenolone and progesterone to their 17-OH derivatives and the conversion of 17-hydroxyprogesterone and 17-hydroxypregnenolone to DHEA and androstenedione via its lyase activity, leading to the synthesis of androgenic steroids in the gonads, adrenals, fat, and tumor stroma. Abiraterone acetate (Zytiga) is a prodrug of abiraterone, a potent and selective Cyp17 inhibitor shown to increase overall survival in patients with castration-resistant prostate cancer.

Gonadotropin-Releasing Hormones

Drugs that target receptors for gonadotropin-releasing hormones (GnRHs) decrease the production of ovarian or testicular hormones. The most widely used agents (leuprolide, goserelin) are agonists of GnRH receptors that cause an immediate increase in gonadotropins and eventually produce castration levels of sex hormones by the desensitization of GnRH receptors. A newer agent, abarelix (Plenaxis), is a GnRH receptor antagonist that immediately decreases GnRHs without the disadvantage of an initial hormone surge.

Vascular Endothelial Growth Factor

The observation by Folkman and colleagues that tumors stimulate blood vessel formation, coupled with the knowledge that angiogenesis is required for tumor growth and metastasis, led to a search for effective inhibitors of this process. Vascular endothelial growth factor (VEGF) is produced by normal and neoplastic cells and regulates angiogenesis. The demonstration by Kim and colleagues that a murine monoclonal antibody against VEGF had preclinical activity led to the development of bevacizumab , a human monoclonal antibody that binds to and inhibits VEGF, preventing its interaction with VEGF receptors (Flt-1 and KDR) that are present on the surface of endothelial cells, thus inhibiting endothelial cell proliferation.

Bevacizumab (Avastin) was first approved for first-line treatment of metastatic colorectal cancer in combination with 5-fluorouracil–based chemotherapy and now has several other indications. Its most common side effects include hypertension, thrombosis, and proteinuria, but asthenia, gastrointestinal perforation, wound dehiscence, hemorrhage, and nephrotic syndrome have been reported, as has been a possible increase in congestive heart failure. Recently, a recombinant fusion protein, ziv-aflibercept (Zaltrap), referred to as a “VEGF trap,” has been developed and approved in combination with FOLFIRI for refractory colorectal cancer. The VEGF trap fuses the extracellular domain of VEGF receptors 1 and 2 with an Fc domain of human IgG1, thereby acting as a soluble receptor for VEGFs.

Drugs Affecting Growth Factor Receptors

Steroid Hormone Receptors

The interaction between steroid hormones and intracellular receptors recruits co-activators and co-repressors to the nuclear transcription complex, leading to transcriptional activation of genes containing specific steroid response elements.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here