Review Article
Biomarkers of cell death applicable to early clinical trials

https://doi.org/10.1016/j.yexcr.2012.03.020Get rights and content

Abstract

The development of biomarkers of cell death to reflect tumor biology and drug-induced response has garnered interest with the development of several classes of drugs aimed at decreasing the cellular threshold for apoptosis and exploiting pre-existing oncogenic stresses. These novel anticancer drugs, directly targeted to the apoptosis regulatory machinery and aimed at abrogating survival signaling pathways, are entering early clinical trials provoking the question of how to monitor their impact on cancer patients. The parallel development of drugs with predictive biomarkers and their incorporation into early clinical trials are anticipated to support the pharmacological audit trail, to speed the development and reduce the attrition rate of novel drugs whose objective is to provoke tumor cell death. Tumor biopsies are an ideal matrix to measure apoptosis, but surrogate less invasive biomarkers such as blood samples and functional imaging are less challenging to acquire clinically. Archetypal and exploratory examples illustrating the importance of biomarkers to drug development are given. This review explores the substantive challenges associated with the validation, deployment, interpretation and utility of biomarkers of cell death and reviews recent advances in their incorporation in preclinical and early clinical trial contexts.

Introduction

A biomarker is “a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention” [1]. Biomarkers are increasingly being incorporated into early clinical trials [2] with the expectation that they can help document the pharmacological audit trail (described in [3]) which briefly involves:

  • Identification of patients most likely to benefit from a specific drug treatment (a predictive biomarker)

  • Demonstration that potentially active drug concentrations are achieved in the patient (a pharmacokinetic biomarker)

  • Demonstration that a drug hits its desired target (proof of mechanism pharmacodynamic biomarker, a POM PD biomarker)

  • Achievement of appropriate drug-induced biological effect (proof of concept pharmacodynamic biomarker, POC PD biomarker)

  • Monitoring clinical response to therapy (a surrogate response biomarker).

The importance of biomarkers in drug development is illustrated by the development of trastuzumab, a monoclonal antibody targeting the HER-2 protein, whose development may have failed had a companion diagnostic (a predictive biomarker) not been used to restrict the trial to patients over-expressing the drug target [4]. Similarly bortezomib, a proteosome inhibitor, achieved its initial license in the treatment of myeloma on the basis of changes in a surrogate response biomarker [5]. The parallel development of drugs with predictive biomarkers that allow patient stratification and PD POM and POC biomarkers to facilitate dose and schedule selection is anticipated to speed up the development and reduce the attrition rate of new drugs in clinical development.

While debate continues about when such biomarkers should be deployed during drug development [6], [7], defining the dose of drug that hits its molecular target and elicits an appropriate tumor response is a key goal prior to embarking on expensive late stage trials. In this review, we examine the challenges and recent advances in the implementation of POC PD biomarkers to assess the optimal drug-induced tumor cell endpoint: cell death.

Section snippets

Biomarkers of cell death to reflect tumor biology and drug response

Increased cell survival is a hallmark of cancer [8], and suppression of apoptosis is important both in the development of the primary tumor and metastasis. Paradoxically, dominant oncogenes such as c-Myc stimulate both proliferation and apoptosis [9]. Despite the over-expression of anti-apoptotic proteins that commonly occurs in human cancers to restrict oncogene-driven cell death [10], more aggressive tumors of a higher histological grade frequently display relatively high levels of apoptosis

Apoptosis biomarkers in clinical trials

The EU regulations for biomarker analysis in clinical trials stipulate that the analysis should be performed to a quality standard in accredited laboratories working to Good Clinical Practice for Laboratories (GCPL, [20]). Thus all biomarker assays should be robust, reliable and reproducible and designed for sufficiently high assay performance for the clinical sample in which they will be measured. Only when these criteria are fulfilled will it be acceptable to use biomarker data from a

Measurement of cell death in tumor biopsies

The classical assessment of apoptosis was based on morphological criteria [21], although this has progressively moved towards biochemical criteria [22]. Measurement of cellular components non-randomly degraded during apoptosis has been applied to tumor biopsies, e.g. the TUNEL assay where labeled bromodeoxyuridine is incorporated in the DNA ends by the action of terminal deoxynucleotidyl transferase [23], [24]. More commonly, morphological assessment by H&E staining alongside

Measurement of cell death biomarkers in peripheral blood mononuclear cells (PBMCs)

Measurement of cell death in PBMCs may be used when biopsies are unavailable. Determining the relationship between death in the surrogate and tumor tissue may be relatively straightforward; for example in patients with lymphoma a Phase II study of bryostatin 1 and vincristine demonstrated an association between death of circulating CD5 + T cells (assessed by Annexin V staining) and tumor cell death [31]. Phase I trials in patients with solid tumors sometimes include PBMCs as a surrogate tissue

Circulating biomarkers of apoptosis

Blood-based molecular tests that can be performed on circulating cancer cells, plasma or serum are less invasive, have greater utility to characterize temporal changes, and are more readily applied to most clinical settings. Serial sampling provides a dynamic picture of apoptosis and real-time view of the individual patient's disease course and response to treatment. This may be particularly helpful if the released cell death biomarker is removed slowly from the circulation so that a cumulative

Integrating preclinical qualification into the design of early clinical trials

The utility of preclinical studies to validate biomarkers and/or determine optimum timing of samples has been demonstrated in the development of several drugs targeting apoptosis pathways. The impact of the aurora kinase inhibitor AZD1152 [29] on apoptosis was evaluated in tumor and the bloodstream in a preclinical in vivo model. In treated animals bearing human colon cancer xenografts (SW620), plasma cCK18 increased 2–3 fold by day 5 compared with controls. Confirmation that the signal was

Circulating tumor cells

The critical issue in the interpretation of the aforementioned circulating biomarkers is the lack of specificity in distinguishing tumor response from host tissue toxicity. The identification, enumeration and molecular characterization of circulating tumor cells (CTCs) offer potential real-time assessment of genotypic and phenotypic features of cancer, avoiding an invasive biopsy or overlapping signal from host toxicity. CTC detection technologies (reviewed in [55]) can be broadly divided into

Functional imaging of tumor cell death

A non-invasive assessment of tumor apoptosis that is gaining momentum is functional imaging. The detection of apoptosis by Annexin V conjugated to a radio-isotope correlated with response in patients with lung cancer and lymphoma receiving chemotherapy [61]. However, challenges in differentiating the drug-induced signal from that of physiological apoptosis and low resolution of the technique meant that only areas of high apoptosis in large tumors were detectable. While progress is being made in

Conclusions

The utility of the majority of apoptosis biomarker data acquired in clinical trials to-date has been limited by the use of unvalidated biomarker assays, with no prospective pre-clinical work to determine the optimum sampling times and uncertain definitions of the magnitude of biomarker signal change considered a significant drug-induced change. If biomarkers are to be used in decision-making for drug development or schedule selection, it is vital that they are measured with validated assays.

References (64)

  • K. Chen et al.

    Positron emission tomography imaging of cancer biology: current status and future prospects

    Semin. Oncol.

    (2011)
  • Biomarkers Definitions Working Group National Institute of Health

    Biomarkers and surrogate endpoints: preferred definitions and conceptual framework

    Clin. Pharmacol. Ther.

    (2001)
  • B.H. Goulart et al.

    Trends in the use and role of biomarkers in phase I oncology trials

    Clin. Cancer Res.

    (2007)
  • T.A. Yap et al.

    Envisioning the future of early anticancer drug development

    Nat. Rev. Cancer

    (2010)
  • M.J. Duffy

    Predictive markers in breast and other cancers: a review

    Clin. Chem.

    (2005)
  • R.C. Kane et al.

    United States Food and Drug Administration approval summary: bortezomib for the treatment of progressive multiple myeloma after one prior therapy

    Clin. Cancer Res.

    (2006)
  • R.H. Glassman et al.

    Biomarkers in early cancer drug development: limited utility

    Clin. Pharmacol. Ther.

    (2009)
  • C.P. Carden et al.

    From darkness to light with biomarkers in early clinical trials of cancer drugs

    Clin. Pharmacol. Ther.

    (2009)
  • A.G. Letai

    Diagnosing and exploiting cancer's addiction to blocks in apoptosis

    Nat. Rev. Cancer

    (2008)
  • M. Parton et al.

    Coordinate expression of apoptosis-associated proteins in human breast cancer before and during chemotherapy

    Clin. Cancer Res.

    (2002)
  • I.B. Weinstein et al.

    Mechanisms of disease: oncogene addiction—a rationale for molecular targeting in cancer therapy

    Nat. Clin. Pract. Oncol.

    (2006)
  • F.L. Rehman et al.

    Synthetic lethal approaches to breast cancer therapy

    Nat. Rev. Clin. Oncol.

    (2010)
  • T.H. Ward et al.

    Biomarkers of apoptosis

    Br. J. Cancer

    (2008)
  • C. Dive et al.

    Drug-target interactions: only the first step in the commitment to a programmed cell death?

    Br. J. Cancer

    (1991)
  • S.H. Kaufmann et al.

    Alterations in the apoptotic machinery and their potential role in anticancer drug resistance

    Oncogene

    (2003)
  • B. Leber et al.

    Drugs targeting bcl-2 family members as an emerging strategy in cancer

    Expert Rev. Mol. Med.

    (2010)
  • J. Cummings et al.

    Biomarker method validation in anticancer drug development

    Br. J. Pharmacol.

    (2008)
  • A.H. Wyllie et al.

    Cellular events in the adrenal cortex following ACTH deprivation

    J. Pathol.

    (1972)
  • G. Kroemer et al.

    Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009

    Cell Death Differ.

    (2009)
  • V. Guarneri et al.

    Phase II, randomized trial of preoperative epirubicin-paclitaxel ± gefitinib with biomarker evaluation in operable breast cancer

    Breast Cancer Res. Treat.

    (2008)
  • L.S. Wang et al.

    Modulation of genetic and epigenetic biomarkers of colorectal cancer in humans by black raspberries: a phase I pilot study

    Clin. Cancer Res.

    (2011)
  • W.G. Tong et al.

    Phase I and pharmacologic study of SNS-032, a potent and selective Cdk2, 7, and 9 inhibitor, in patients with advanced chronic lymphocytic leukemia and multiple myeloma

    J. Clin. Oncol.

    (2010)
  • Cited by (0)

    1

    These authors contributed equally.

    View full text