Cancer Pharmacology


Summary of Key Points

  • Cancer pharmacology encompasses the spectrum of therapeutic issues from everyday clinical treatment to the earliest stages of new drug discovery.

  • Translational research has led to more sophisticated identification of targets that are relevant for therapy, and clinical success has attracted wide corporate involvement.

  • Regulatory flexibility has made new therapies available to patients at an unprecedented rate and has provided a variety of options for developmental strategies.

  • The era of oral drugs for cancer treatment is well underway.

  • Once the drug is selected to match the tumor, drug delivery must provide adequate systemic exposure for tumors while minimizing toxicity and adjusting for concomitant therapy.

Cancer pharmacology offers an integrated body of practical knowledge that is valuable for the full range of activities related to cancer therapeutics, from everyday clinical treatment decisions, to assisting clinical trial design in drug development, to discovery and evaluation of compounds with potential for advancing to the clinic.

One of the most obvious metrics for the importance of cancer pharmacology is the rapid and continuing expansion in the number of drugs available to treat cancer. More than 200 agents have been approved for marketing in oncology, including 51 that were approved by the US Food and Drug Administration (FDA) from 2011 to 2015. Notably, 15 were approved in 2015 ( Table 25.1 ). As a point of reference, at the first meeting of the American Society of Clinical Oncology (ASCO) in 1964, there were only 12 drugs that had been previously approved for marketing in oncology.

Table 25.1
Cancer Drugs Approved by the Food and Drug Administration in 2015 a
Drug Route Type
Daratumumab Intravenous Monoclonal antibody
Dinutuximab Intravenous Monoclonal antibody
Elotuzumab Intravenous Monoclonal antibody
Necitumumab Intravenous Monoclonal antibody
Alectinib Oral Small synthetic molecule
Cobimetinib Oral Small synthetic molecule
Ixazomib Oral Small synthetic molecule
Lenvatinib Oral Small synthetic molecule
Osimertinib Oral Small synthetic molecule
Panobinostat Oral Small synthetic molecule
Palbociclib Oral Small synthetic molecule
Sonidegib Oral Small synthetic molecule
Trabectedin Intravenous Small synthetic molecule
Trifluridine/tipiracil Oral Small synthetic molecule
Uridine triacetate Oral Small synthetic molecule

a Of the 15 new drugs, 4 are monoclonal antibodies delivered intravenously. Of the 11 small molecules, 10 are delivered orally.

When ASCO was founded, the National Cancer Institute (NCI) was the primary source for new cancer drugs, and the NCI continues to conduct an extensive program of cancer drug development. NCI also collaborates extensively with industry and other organizations. Over the past 25 years, the pharmaceutical industry has dramatically increased the resources invested in cancer pharmacology. Various nonprofit organizations also are engaged in the development of cancer drugs, but the private sector is the largest contributor to the effort to bring new treatments to patients with cancer.

Beyond the impressive increase in the number of drugs available to treat cancer, other factors are fundamentally changing the way that oncologists and drug developers discover, develop, and prescribe drugs for treatment of patients with cancer. In most cases, there is a strong trend toward narrow indications, as the broad histopathologic categories of cancer have become fractionated into many subtypes. There is also a countertrend: seeking drug approvals based on the target for a drug, rather than the tissue site. In addition, because effective therapy is more widely available for first-line treatment of many cancers, initial approvals are further stratified by the extent of prior treatment.

The approval of imatinib in 2001 heralded several of the most dramatic changes in cancer therapy, foreshadowing entirely new paradigms that became commonplace during the next 15 years: molecular targeting, diagnostic tests associated with selection of therapy, and the beginning of the shift from intravenous to oral drug delivery in patients with cancer.

Although this chapter focuses on small, synthetic molecules, one of the most exciting developments in cancer pharmacology is the successful transition of antibodies from the research stage to full integration with small molecules in the everyday treatment of patients. There is also a role for toxins that are linked to antibodies, which can help to guide the toxin to the tumor (see Chapter 30 ). Although very different in terms of their size and manufacturing techniques, the principles of development for these new therapeutic classes have the same goals as for small molecules—for example, modulation of specific molecular targets and linkage to a diagnostic test for that target. Trastuzumab embodies all of these elements, including its extension into the realm of antibody-drug conjugates with the 2013 FDA approval of ado-trastuzumab emtansine.

The rapid increase in availability of new drugs and new data on how to optimally use drugs, including the ability to avoid drug-drug and drug-food interactions, is an enormously valuable situation for patients with cancer. How will oncologists keep up with the avalanche of information for all of these issues? Everything an oncologist needs to know can no longer be squeezed into a small book that fits in the pocket of the physician's white coat. Electronic versions of textbooks and centralized electronic databases help provide the intricate pieces of data. In this chapter the focus is on the principles of cancer pharmacology and its ability to help categorize data and organize it into useful information.

Fundamental Science

The two major disciplines within cancer pharmacology are drug actions (pharmacodynamics [PD]) and drug delivery (pharmacokinetics [PK]). Both of these areas are highly dependent on knowledge derived from pharmacogenetics and pharmacogenomics.

In cancer pharmacology the primary basis for success or failure of drug therapy is the ability to find a treatment that is matched to the intrinsic sensitivity of the tumor. No therapeutic benefit will be achieved if the target for drug action is not present in the tumor. However, the perfect match between target and therapy can still fail if adequate systemic exposure of drug is not delivered to the tumor.

Systemic exposure is pivotal. For a responsive tumor, higher systemic exposure can be associated with greater benefit but also more adverse effects in normal tissue. Lower systemic exposure can have lower toxicity in normal tissues, but at the risk of reduced effect on the tumor. The therapeutic index for a cancer drug is the ratio of its antitumor activity versus the adverse effects on normal tissues.

The principles that connect dose, systemic exposure, and toxicity have been demonstrated regularly for normal tissues in the body, but the relationships of systemic exposure to antitumor effect are far more difficult to discern. Understanding the linkage between drug delivery and drug actions continues to be a major challenge in cancer pharmacology.

Principles of Cancer Drug Action

Genetics is a fundamental part of the study of cancer research (see Chapter 1 ). The range of applications for genetics in cancer includes hereditary predisposition and interactions of heredity with nonhereditary factors (see Chapter 13 ).

In cancer pharmacology, molecular targets that could be relevant to therapy have received the most intense emphasis of pharmacogenomics. This focus includes unique proteins coded by translocations, differences in gene expression between tumors and host tissues, and mutations in sequences. The identification of the most attractive molecular targets and the ability to monitor target engagement during therapy are cornerstones for customized treatment.

As described in Chapter 26 and elsewhere (e.g., Chapter 8 ), our understanding of the physiology and molecular biology of tumors has provided a wealth of potential targets for anticancer therapy. Although the relative intensity has magnified, the concept of molecular targeting and linkage to diagnostics for selection and monitoring of individual patients has a long history. The selection of hormonal therapies for patients whose tumors express the estrogen receptor was among the first successful uses of molecular targeting.

The current era of targeting began with approval of trastuzumab (1998) only in patients with Her2-positive tumors and approval of imatinib (2001) only in patients with chronic myelogenous leukemia (CML) that is Philadelphia chromosome positive. These early successes fueled emphasis on other targets. ALK and BRAF are among the many descendants of the approach. In 2017, the target became more pivotal with the approval for pembrolizumab for treatment of tumors with tumors that demonstrate mismatch repair or microsatellite instability, regardless of tumor's site of origin.

In addition to the obvious attraction of matching drugs to the molecular characteristics of the tumor, the therapeutic index can also be improved by examining host tissues. In his last major research publication, Dr. Abeloff was a leader in a large multicenter study of the pharmacogenetics of cyclophosphamide. The goal was to determine whether the germline DNA of patients altered the toxicity profile of cyclophosphamide. The researchers found a subgroup of women who had variant GSTP1 alleles that were associated with less susceptibility to adverse hematologic toxicity in regimens containing cyclophosphamide. Subsequent work by large cooperative groups in the United Kingdom for paclitaxel in breast cancer and in the United States for docetaxel in prostate cancer found other links between germline DNA and sensory neuropathy.

Although these studies were only hypothesis generating, the examples illustrate the possible ways that the therapeutic index for a drug can be improved with examination of host tissues. Later in this chapter, in the discussion of drug delivery, the pharmacogenetics of metabolism and cellular membrane transporter cell membranes in host tissues are discussed.

Combinations of Drugs

As described in the various chapters relating to specific malignancies in Part III of this book, it is rare for a drug to be used as a single agent. The development of combinations would be an extensive topic in itself. Box 25.1 provides a set of points to consider for combinations. Within a combination chemotherapy regimen, the drugs are intended to interact at the pharmacodynamic level. Originally, a major strategy was to combine cancer drugs with nonoverlapping toxicities. Increasingly, more detailed knowledge regarding pathways for cancer drugs permits the design and evaluation of combination strategies that target parallel and/or sequential pathways.

Box 25.1
Strategic Factors for Drug Combinations

  • Rationale for target-based combinations

  • Addition of new drug to established regimen

  • Systematic evaluation of all combinations

  • Translation of findings into clinical trials

Most combinations are based on a preexisting scientific rationale or by addition of a new agent to an established regimen. However, it would be presumptuous to assume that all possibilities are already understood. One tool to expand the scope of hypothesis generation is the systematic study of all cancer drugs in combination with one another or in combination with approved agents outside oncology, or the testing of every investigational agent versus all approved cancer drugs. Although this screening approach begins as an empiric exercise, the successful combinations generate challenges for explaining the underlying mechanisms of action. The focus on drugs that are already approved provides a potentially fast route to clinical implementation.

Molecular Imaging of Cancer Drug Action

With the widespread availability of fluorine-18 fluorodeoxyglucose ( 18 F-FDG), a consensus was building in the late 1990s that positron emission tomography (PET) would emerge as a tool for functional assessment, which would complement anatomic imaging modalities. Since then, FDG has been highly successful as a general probe in many tumor types, providing additional information to help separate malignant from benign lesions and to monitor response to therapy.

FDG is now an approved agent for imaging. Its success has spurred interest in the development of other probes for PET imaging that are currently in the investigational stage of clinical evaluation, including fluorine-18 fluorothymidine ( 18 F-fluorothymidine; FLT). Liu et al. demonstrated the potential for FLT in a patient with uterine cancer. At baseline, the FLT image shows major uptake by the tumor, indicative of active proliferation. During treatment with sunitinib, uptake of FLT in the tumor decreased, which was interpreted as reduced proliferation. The image following the withdrawal of treatment exhibited a flare—that is, intensity that rebounded above the baseline value. Anatomic imaging by computed tomography (CT) readily identified the tumor and could monitor tumor size over the subsequent weeks or months, but FLT provided the ability to follow molecular pathways in real time.

Cancer Drug Delivery: Systemic Exposure

Once the appropriate drug has been chosen, the most important everyday question in clinical cancer pharmacology is the selection of an appropriate dose. The standard dose determined previously in population studies is a starting point for consideration, but what factors could make this dose too high or too low for individual patients? For many cancer drugs, evidence-based advice is not available. In some cases, guidance for dose adjustment is available based on prior courses of treatment or age.

Increasingly, cancer drugs are approved with information about dosage adjustment based on renal function testing, food intake, and the concurrent use of other drugs. All of these factors can produce variability in systemic exposure. Thus the delivery of drugs to both the tumor and normal tissues is affected. In addition to empiric data from clinical investigations for specific drugs, what are the principles that underlie these decisions based on drug delivery?

Clearance is at the top of the list for drug delivery parameters in Box 25.2 because it is the underlying factor with the greatest impact on dose adjustment in cancer drug delivery. As stated in Box 25.3 , clearance is the summation of all mechanisms for a drug to be removed from the systemic circulation, sometimes called total body clearance. Several specific applications of clearance for dosage adjustment are covered in the next few sections.

Box 25.2
Drug Delivery Concepts

  • Clearance (dose adjustment)

    • Impaired organ function

    • Drug-drug interactions

    • Metabolic

    • Transport

    • Pharmacogenetic factors

  • Bioavailability

    • How much drug reaches systemic circulation

    • Includes food effects

  • Half-life (dose interval)

  • Volume of distribution, protein binding

Box 25.3
Role of Clearance in Dosage Adjustment

  • Clearance: summation of all routes of drug elimination from the body―renal, hepatic, other

  • Primary application: guide dose adjustment

    • Low clearance → high exposure (consider reduced dose)

    • High clearance → low exposure (consider increased dose)

Bioavailability, which is the second concept listed in Box 25.2 , is a measure of how much drug enters the systemic circulation. Because of the shift toward oral therapy, bioavailability has risen sharply in importance. This concept is described further in the section Oral Cancer Drugs.

Half-life was previously the most discussed parameter for cancer drug delivery. However, its role is primarily in the development stage, to help explore frequency of administration. Once a drug has been approved for clinical practice, oncologists do not have a frequent need to know the specific value for half-life of a cancer drug. Other drug delivery parameters such as volume of distribution or protein binding can have value in specific situations, but only on rare occasions.

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