Pharmacogenomics in acute lymphoblastic leukemia

https://doi.org/10.1016/j.beha.2017.07.007Get rights and content

Abstract

Pharmacogenomics is a fast-growing field of personalized medicine using a patient's genomic profile to determine drug disposition or response to drug therapy, in order to develop safer and more effective pharmacotherapy. Childhood acute lymphoblastic leukemia (ALL), being the most common malignancy in childhood, which is treated with uniform and standardized clinical trials, is remarkably poised for pharmacogenomic studies. In the last decade, unbiased genome-wide association studies have identified multiple germline risk factors that strongly modify host response to drug therapy. Some of these genomic associations (e.g. TPMT, NUDT15 and mercaptopurine dosing) have accumulated a significant level of evidence on their clinical utility such that they are warranted as routine clinical tests to guide modification of treatment. Most of these germline associations however, have not yet reached such actionability. Insights have also been gathered on germline factors that affect host susceptibility to adverse effects of antileukemic agents (eg, vincristine, asparaginase, methotrexate). Further large-scale studies are required, along with the assimilation of both germline and somatic variants, to precisely predict host drug response and drug toxicities, with the eventual aim of executing genomic-based precision-pharmacotherapy in the treatment of ALL.

Introduction

The study of pharmacogenomics is a fast-growing research field, combining human genetics and molecular pharmacology, to determine how inter-individual genetic variations contribute to a patient's interaction with or response to drugs [1]. By using a patient's genomic profile to elucidate functionally relevant genomic determinants for drug disposition or response to drug therapy, we can aim to develop safer and more effective pharmacotherapy [2]. The idea is that the choice of treatment can be evaluated by weighing the probability of cure against the probability of adverse effects based on genetic characteristics. Host genomic profiles may also indicate drugs with little importance for cure but which are associated with a high risk for side effects for specific patients, allowing for better tailored therapy.

With rapid advances in genomic profiling technology, we are now able to efficiently examine variants across the entire genome in genome wide association studies (GWAS), with testing and analyzing a few million genetic markers per patient to determine the association of these variants with a particular phenotype of interest [3]. These large-scale studies have provided invaluable insight into the genetic basis of drug efficacy and toxicity during leukemia treatment.

Due to the exceedingly high number of variants analyzed in GWAS, the required level of significance for association between these variants and any particular phenotype is generally set at a much more stringent level of significance (P < 5 × 10−7), compared to the usual level of 0.05. Thus, it is expected that only genomic variations with relatively large effect sizes are detected in these studies, especially when the sample size is modest [4].

The majority of pharmacogenomics studies in leukemia are performed in childhood acute lymphoblastic leukemia (ALL), as ALL is the most common childhood cancer comprising up to 25% of all childhood malignancies and also a model disease for pharmacogenomics research, for many unique reasons. The translation of theis knowledge into new chemotherapeutic paradigms can serve as good models for optimizing treatment of other malignancies [5].

Firstly, childhood ALL generally has very standardized treatment protocols and most children under treatment are enrolled on prospective and uniform protocols where important data including presenting features, medication history, treatment outcome, and adverse events are systematically recorded.

Second, combination chemotherapy is highly efficacious in childhood ALL with a large majority of the patients being cured on pharmacotherapy alone. With medications playing such a large role in treatment, one can infer that pharmacogenomics would significantly influence the outcome of these patients. Furthermore, these medications operate on narrow ranges of therapeutic indices, hence it is imperative for us to understand the underlying genetic basis of antileukemic response and adverse effects, so that we can carefully navigate patients into the optimal therapeutic range of these drugs.

Most contemporary childhood ALL protocols utilize genomics of the ALL blasts to risk-stratify patients in order to decide the intensity of treatment. Hence, clinicians are already used to adjusting treatment regimens according to patient genomics.

Finally, many medications commonly used to treat childhood ALL are also used to treat many other non-malignant disorders. Genetic variants associated with drug effects, particularly adverse events, may have relevance for these other conditions as well. Knowledge gleaned from pharmacogenomics of these anti-leukemic agents can aid physicians in using the same class of drugs more effectively and appropriately for other conditions [1].

In cancer, both the somatically acquired genomic features of the malignancy and the inherited genomic variation of the host have the potential to influence efficacy of therapy. In this review article, we will focus primarily on germline variants which influence outcome of ALL. These variants affect the efficacy of antileukemic treatment through effects on response to treatment (as evidenced by MRD), drug sensitivity and relapse, as well as adverse drug effects [1].

Section snippets

Pharmacogenomics of ALL treatment outcome

The ultimate outcome of ALL therapy is governed by a wide variety of factors, including the patient's response to antileukemic agents, interactions between leukemia cells and its microenvironment, as well as the biology of leukemia cells itself. Via a multitude of both genome wide interrogation studies and candidate gene studies, there is increasing discovery of inherited germline variations associated with both early treatment response (minimal residual disease, MRD) and the risk of ALL

Pharmacogenomics of adverse effects of ALL therapy

With the exception of thiopurines, pharmacogenetic variants for other toxicity phenotypes have not been well validated. This is perhaps due to the fact that it is difficult to control for variability in drug exposure in studies to elucidate the genetic basis for adverse effects. Subtle differences in drug exposure can already have significant impact on the severity of toxicity, and substantial variability exists amongst ALL treatment regimens. These factors likely contribute to the relative

Conclusion

Taken together, both candidate-gene and genome-wide studies have identified inherited genetic variations related to interpatient variability in ALL treatment outcomes and toxicity. However, the extent of interplay between inherited germline variants and somatic ALL tumor genetic factors and its impact on therapy response is not entirely clear. Currently, other than well established genes such as TPMT or NUDT15, there are relatively few germline genomic associations that have the required level

Conflict of interest

The authors declare that there are no conflicts of interest.

Practice points

  • Pharmacogenomics study how a patient's genetic make-up influences his/her response to drugs. Pharmacogenomics-guided precision medicine contributed significantly to the success of childhood ALL therapy.

  • Multiple genome wide association studies have identified multiple germline risk factors related to efficacy and/or toxicities of ALL therapy

  • Germline genomic variants in the TPMT, IL15, GATA3, PYGL, PDE4B genes have been linked to treatment response in children with ALL

  • Germline genomic variants

Research agenda

  • Integrated analyses including both germline and somatic genetic variations to provide comprehensive characterization of genetic risk factors for ALL relapse to help individualise therapy

References (55)

  • J.P. Cai et al.

    Mouse MTH2 protein which prevents mutations caused by 8-oxoguanine nucleotides

    Biochem Biophys Res Commun

    (2003)
  • Y. Takagi et al.

    Human MTH3 (NUDT18) protein hydrolyzes oxidized forms of guanosine and deoxyguanosine diphosphates: comparison with MTH1 and MTH2

    J Biol Chem

    (2012)
  • R. Pacheco et al.

    Role of glutamate on T-cell mediated immunity

    J Neuroimmunol

    (2007 Apr)
  • J.D. Kawedia et al.

    Pharmacokinetic, pharmacodynamic, and pharmacogenetic determinants of osteonecrosis in children with acute lymphoblastic leukemia

    Blood

    (2011)
  • B. Diouf et al.

    Genome-wide association analyses identify susceptibility loci for vincristine-induced peripheral neuropathy in children with acute lymphoblastic leukemia

    Blood

    (2013)
  • S. Radtke et al.

    Germline genetic variations in methotrexate candidate genes are associated with pharmacokinetics, toxicity, and outcome in childhood acute lymphoblastic leukemia

    Blood

    (2013)
  • L.B. Ramsey et al.

    Genome-wide study of methotrexate clearance replicates SLCO1B1

    Blood

    (2013)
  • D. French et al.

    Acquired variation outweighs inherited variation in whole genome analysis of methotrexate polyglutamate accumulation in leukemia

    Blood

    (2009)
  • M.V. Relling et al.

    Pharmacogenomics of acute lymphoid leukemia: new insights into treatment toxicity and efficacy

    Hematol Am Soc Hematol Educ Program

    (2013)
  • L. Kager

    Pharmacogenomics to improve childhood acute lymphoblastic leukaemia therapy

    Mag Eur Med Oncol

    (2009)
  • M.I. McCarthy et al.

    Genome-wide association studies for complex traits: consensus, uncertainty and challenges

    Nat Rev Genet

    (2008)
  • P.C. Sham et al.

    Statistical power and significance testing in large-scale genetic studies

    Nat Rev Genet

    (2014)
  • Meyling H. Cheok et al.

    Acute lymphoblastic leukaemia: a model for the pharmacogenomics of cancer therapy

    Nat Rev Cancer

    (2006 Feb)
  • L. Holmfeldt et al.

    The genomic landscape of hypodiploid acute lymphoblastic leukemia

    Nat Genet

    (2013)
  • J. Zhang et al.

    The genetic basis of early T-cell precursor acute lymphoblastic leukaemia

    Nature

    (2012)
  • M.V. Relling et al.

    Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S- methyltransferase gene locus

    J Natl Cancer Inst

    (1999)
  • M. Stanulla et al.

    Thiopurine methyltransferase (TPMT) genotype and early treatment response to mercaptopurine in childhood acute lymphoblastic leukemia

    JAMA

    (2005)
  • Cited by (24)

    • Insights into S-adenosyl-L-methionine (SAM)-dependent methyltransferase related diseases and genetic polymorphisms

      2021, Mutation Research - Reviews in Mutation Research
      Citation Excerpt :

      As the number of leukemia cells increases in the blood and bone marrow of ALL patients, healthy white blood cells, red blood cells and platelets are crowded out, leading to infection, anemia and bleeding. The purine analogues mercaptopurine and thioguanine are drugs that have been used as cytotoxic agents in the treatment of leukemias for more than 50 years [151]. Because the therapeutic window between toxicity and efficacy is narrow, thiopurine drugs can potentially have life-threatening toxicity [152].

    • Pharmacogenomic mechanisms of drug toxicity

      2021, Atkinson's Principles of Clinical Pharmacology
    • The global role of biotechnology for non communicable disorders

      2018, Journal of Biotechnology
      Citation Excerpt :

      For instance, trastuzumab (Hereceptin), is a monoclonal antibody (see below) that works only in women with breast cancers having a particular genetic profile that leads to overproduction of the HER2 (Ahmed et al., 2015). Genetic testing is also recommended before administering chemotherapy based on mercaptopurine (Purinethol) to patients with acute lymphoblastic leukemia, as some genetic variants interfere with the ability to process the drug and can increase the risk of infection, unless the dose is adjusted according to the genetic profile (Lee and Yang, 2017). A similar dose tailoring approach, has been proposed for the blood thinning drug warfarin (Coumadin), which is included in the therapeutic regimen for various NCDs, although more research is needed to conclusively determine whether warfarin dosing based on the genetic information is better than the current trial-and-error approach (Tseng et al., 2018).

    • Guest editorial: recent progress in pediatric leukemia

      2023, International Journal of Hematology
    View all citing articles on Scopus
    View full text