Elsevier

Blood Reviews

Volume 43, September 2020, 100653
Blood Reviews

Review
Peripheral neuropathy in hematologic malignancies – Past, present and future

https://doi.org/10.1016/j.blre.2020.100653Get rights and content

Abstract

Neurotoxic treatments (including proteasome inhibitors, immunomodulatory drugs and vinca-alkaloids) are often used in the treatment of hematologic malignancy. Peripheral neuropathy can be part of a paraneoplastic syndrome accompanying the disease but more commonly is a consequence of treatment with neurotoxic therapies, and produces sensory, motor, autonomic nerve dysfunction or a combination, leading to pain, loss of sensation and functional disability. This review provides an update on peripheral neuropathy in hematologic malignancy, including risk factors, mechanisms and treatment options. We examine the clinical features and risk factors for peripheral neuropathy following bortezomib, thalidomide, brentuximab vedotin and vinca alkaloid treatment, as well as related compounds. We review the current data on pharmacogenetic risk factors for the development of toxicity and highlight areas of future research.

Introduction

Peripheral neuropathy (PN) commonly affects the lives of many patients with hematologic malignancies. Although the etiology can be disease-related, it is most often produced as a treatment-related side effect. Neurotoxic treatments (including proteasome inhibitors, immunomodulatory drugs and vinca-alkaloids) are often used in the treatment of multiple myeloma, lymphomas and leukemias (Table 1). PN occurring during anti-neoplastic treatment can lead to dose reduction, delay or treatment cessation, while persisting symptoms post-treatment can adversely impact patients’ quality of life [1,2].

PN produces sensory, motor, autonomic nerve dysfunction, or combinations, leading to pain, loss of sensation, functional disability and increased falls risk [3]. Treatment-induced PN is most consistently assessed using the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) Neuropathy Sensory subscale. This outcome measure is a clinician based grading scale that scores a patient’s symptoms from grade 0–4 (no symptoms to life threatening symptoms). However, the NCI-CTCAE has been known to under report symptom severity [4], while also lacking interobserver reliability [5] and responsiveness [6]. Despite these shortcomings, the NCI-CTCAE grading scale persists as the most commonly used measure of PN in clinical trials. This feature is reflected throughout this review with the incidence reported using this outcome measure (with mild symptoms denoted grade 1, moderate symptoms grade 2 and severe symptoms grade 3).

With the survival rates of hematologic malignancies steadily increasing [7], a research emphasis on treatment-residual side effects is necessary to ensure cancer survivors maintain high quality of life after completion of cancer treatment. The aim of this review is to provide an update of neurotoxic treatments used in hematologic malignancies, mechanisms producing PN in the clinical setting and treatment and rehabilitative strategies.

Section snippets

Search strategy and quality grading

The search strategy identified articles written in English through searches of the National Center for Biotechnology Information/PubMed database, the Cochrane Library and clinical trial registries using search terms ‘neuropathy’ and ‘neurotoxicity’ with multiple terms including ‘bortezomib’, ‘carfilzomib’, ‘ixazomib’, ‘proteasome inhibitor’, ‘thalidomide’, ‘lenalidomide’, ‘pomalidomide’, ‘immunomodulatory drugs’, ‘vincristine’, ‘vinca’, ‘vinca-alkaloids’, ‘brentuxumab vedotin’, ‘vedotin’,

Disease-related neuropathy

The etiology of PN experienced by patients with hematologic malignancies can be disease-related (Table 3). Multiple myeloma and Waldenström macroglobulinemia are associated with paraproteinemic PN [9,10]. In patients with treatment-naïve multiple myeloma, the prevalence of pre-existing PN ranges from 7.2% to 54% [11,12] with the variance largely depending on outcome measures used. Further studies have suggested more than 80% of pre-treated myeloma patients demonstrate subclinical sensory

Bortezomib

Proteasome inhibitors are actively used in the treatment of multiple myeloma, inducing apoptosis of cancer cells [17] by inhibiting the 26S proteasome, disrupting protein regulation and preventing proteasomal degradation of ubiquitinated proteins in cancer cells [18]. Bortezomib is the first generation proteasome inhibitor approved for clinical use and is effective for the treatment of both newly diagnosed and relapsed or refractory multiple myeloma [19,20]. PN is a common toxicity of

Immunomodulatory drugs

Immunomodulatory drugs are commonly used in the treatment of multiple myeloma. Thalidomide is a synthetic derivative of glutamic acid with the ability to inhibit tumor necrosis factor alpha (TNFα) production [54], and also possess anti-angiogenic [55] and immunomodulatory properties [56]. Lenalidomide and pomalidomide were synthesised with the purpose of increasing efficacy (TNFα inhibition capability) [57] while minimising the toxicities associated with thalidomide.

Vinca-alkaloids

Vinca-alkaloids are a class of chemotherapy derived from the periwinkle plant, and are commonly used for the treatment of Hodgkin and non-Hodgkin lymphomas [77]. The four main vinca-alkaloids in clinical use are vincristine, vinblastine, vinorelbine and vindesine.

Vincristine is the most neurotoxic of the vinca-alkaloids, resulting in PN being a prominent dose-limiting side effect. It induces a distal sensory PN in 18%-70% of adult patients [78,79]. Motor involvement can also occur, manifesting

Brentuximab vedotin

Brentuximab vedotin is an antibody-drug conjugate consisting of a specific antibody targeting CD30, and a microtubule disrupting agent monomethyl auristatin E (MMAE) linked through a protease-cleavable linker [100]. It has demonstrated efficacy across several CD30-expressing lymphomas including Hodgkin lymphoma, systemic anaplastic large cell lymphoma and CD-30 positive peripheral T-cell lymphoma [101,102]. PN is a prominent side effect of brentuximab vedotin, typically presenting as a sensory

Platinum

Platinum compounds including cisplatin and oxaliplatin are commonly associated with PN and utilised across a range of solid tumors. In the treatment of solid tumors, cisplatin-induced PN is strongly associated with cumulative doses of >300 mg/m2 [3,116]. In the setting of hematologic malignancies, there is a lack of specific information regarding PN in association with platinum treatment. Cumulative cisplatin doses in hematologic cancer regimens are often lower than in solid tumours and

Neuropathy outcomes post-treatment

The recovery and reversibility of PN post-treatment varies across neurotoxic treatments used in hematologic malignancies. BIPN is commonly described as reversible in the majority of patients. Typically, PN resolves or improves in a substantial majority of patients, as assessed by conventional grading scales. One series demonstrated that 88% of PN events completely resolved in 2.4 months, with 94% resolving to at least a grade 1 in a median of 2.6 months [120]. Another study found that 79% of PN

Role of genetic risk factors

The search for genetic risk factors for treatment related PN has provided insights into the potential mechanisms underlying the development of peripheral nervous system dysfunction. Overall, single nucleotide polymorphisms (SNPs) in genes associated with drug transport and efflux, neural development and repair, microtubule systems and genes associated with inherited peripheral neuropathies have been linked to treatment related PN in hematologic malignancies. However, there remains little

Current options in treatment and prevention

Although treatment-induced PN remain a common side effect among numerous hematologic malignancy treatments (Table 5), there remains a lack of strategies adopted for its treatment and prevention. Despite a number of clinical trials currently investigating possible treatment and prevention interventions (Table 6), recommendations are limited to regular monitoring with neurologic evaluation and dose modification or treatment cessation to prevent severe and permanent nerve damage [18]. In

Conclusions and future directions

Treatment related peripheral neuropathy is a common side effect in the treatment of hematologic malignancies. Despite the development of newer and novel analogues which are associated with reduced toxicity, PN remains an important consideration in managing patient quality of life and maximizing function. Improved understanding of the mechanistic bases underlying neurotoxicity will assist in the development of targeted neuroprotective approaches. Standardized and objective assessment strategies

Practice points

  • Peripheral neuropathy (PN) remains a common toxicity associated with treatment for hematologic malignancy.

  • Detailed clinical assessment (including baseline and during treatment) is required to monitor the development of toxicity and enable implementation of dose reduction guidelines.

  • Improved assessment tools, including use of objective measures of PN and patient-reported outcomes, are required in clinical studies and clinical trials of neuroprotection.

  • Further collaboration with neurologists and

Research agenda

  • Examination of mechanisms for individual agents is required to guide development of neuroprotective therapy.

  • Detailed studies with objective assessment tools are needed to clarify PN risk, particularly with newer agents.

  • The development of better assessment tools to quantify PN in the clinical setting and a standardized set of clinical trial outcome measures.

  • Better understanding of risk factors, including genetic risk factors, which may lead to individual patients having a higher risk of severe

Funding

This work was supported by the National Health and Medical Research Council of Australia [#1080521, 1148595] and the Cancer Institute of New South Wales [14/TPG/1-05].

Declaration of competing interest

Professor Lazarus is a consultant and promotional speaker for Seattle Genetics and Bristol-Myers Squibb and a consultant for Celgene Corporation.

References (175)

  • M.A. Dimopoulos et al.

    Carfilzomib and dexamethasone versus bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma (ENDEAVOR): a randomised, phase 3, open-label, multicentre study

    Lancet Oncol

    (2016)
  • J.D. Federspiel et al.

    Specificity of protein covalent modification by the electrophilic proteasome inhibitor carfilzomib in human cells

    Mol Cell Proteomics

    (2016)
  • K. Strasser et al.

    Thalidomide treatment in multiple myeloma

    Blood Rev

    (2002)
  • I.E. Fahdi et al.

    Bradycardia during therapy for multiple myeloma with thalidomide

    Am J Cardiol

    (2004)
  • S. Zweegman et al.

    Melphalan, prednisone, and lenalidomide versus melphalan, prednisone, and thalidomide in untreated multiple myeloma

    Blood

    (2016)
  • M.A. Dimopoulos et al.

    Safety and efficacy of pomalidomide plus low-dose dexamethasone in STRATUS (MM-010): a phase 3b study in refractory multiple myeloma

    Blood

    (2016)
  • H. Zavala et al.

    Vincristine-induced vocal cord paresis and paralysis in children

    Int J Pediatr Otorhinolaryngol

    (2019)
  • S. Keller et al.

    Neurotoxicity of stem cell mobilization chemotherapy with vinorelbine in myeloma patients after bortezomib treatment

    Leuk Res

    (2015)
  • L. Liu et al.

    Vindesine-induced neuropathy mimicking Guillain-Barre syndrome

    Leuk Res

    (2009)
  • S.M. Ansell

    Brentuximab vedotin

    Blood

    (2014)
  • V. Chaudhry et al.

    Characteristics of bortezomib- and thalidomide-induced peripheral neuropathy

    J Peripher Nerv Syst

    (2008)
  • F. Mols et al.

    Chemotherapy-induced peripheral neuropathy and its association with quality of life: a systematic review

    Support Care Cancer

    (2014)
  • S.B. Park et al.

    Chemotherapy-induced peripheral neurotoxicity: a critical analysis

    CA Cancer J Clin

    (2013)
  • K.A. Griffith et al.

    Measures of chemotherapy-induced peripheral neuropathy: a systematic review of psychometric properties

    J Peripher Nerv Syst

    (2010)
  • R.L. Siegel et al.

    Cancer statistics, 2019

    CA Cancer J Clin

    (2019)
  • M.H. Ebell et al.

    Strength of recommendation taxonomy (SORT): a patient-centered approach to grading evidence in the medical literature

    Am Fam Physician

    (2004)
  • M.L. Mauermann

    Neurologic complications of lymphoma, leukemia, and paraproteinemias

    Continuum (Minneapolis, Minn)

    (2017)
  • C.J. Klein et al.

    The neuropathies of Waldenstrom’s macroglobulinemia (WM) and IgM-MGUS

    Can J Neurol Sci

    (2011)
  • C. Leone et al.

    An observational study assessing peripheral neuropathy related to multiple myeloma

    Neurol Sci

    (2016)
  • P.G. Richardson et al.

    Single-agent bortezomib in previously untreated multiple myeloma: efficacy, characterization of peripheral neuropathy, and molecular correlations with response and neuropathy

    J Clin Oncol

    (2009)
  • A.K. Kosturakis et al.

    Subclinical peripheral neuropathy in patients with multiple myeloma before chemotherapy is correlated with decreased fingertip innervation density

    J Clin Oncol

    (2014)
  • C. Briani et al.

    Peripheral neuropathies in chronic lymphocytic leukemia: a single center experience on 816 patients

    Haematologica

    (2017)
  • J.J. Kelly et al.

    Lymphoma and peripheral neuropathy: a clinical review

    Muscle Nerve

    (2005)
  • K. Viala et al.

    Neuropathy in lymphoma: a relationship between the pattern of neuropathy, type of lymphoma and prognosis?

    J Neurol Neurosurg Psychiatry

    (2008)
  • P.M. Voorhees et al.

    The proteasome as a target for cancer therapy

    Clin Cancer Res

    (2003)
  • P.G. Richardson et al.

    Management of treatment-emergent peripheral neuropathy in multiple myeloma

    Leukemia

    (2012)
  • J.F. San Miguel et al.

    Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma

    N Engl J Med

    (2008)
  • P.G. Richardson et al.

    Bortezomib or high-dose dexamethasone for relapsed multiple myeloma

    N Engl J Med

    (2005)
  • K. Scott et al.

    Bortezomib for the treatment of multiple myeloma

    Cochrane Database Syst Rev

    (2016)
  • P.G. Richardson et al.

    Complications of multiple myeloma therapy, part 1: risk reduction and management of peripheral neuropathy and asthenia

    J Natl Compr Canc Netw

    (2010)
  • A. Stratogianni et al.

    Bortezomib-induced severe autonomic neuropathy

    Clin Auton Res

    (2012)
  • R. Velasco et al.

    Bortezomib and other proteosome inhibitors-induced peripheral neurotoxicity: from pathogenesis to treatment

    J Peripher Nerv Syst

    (2019)
  • I. Casafont et al.

    Bortezomib induces the formation of nuclear poly(A) RNA granules enriched in Sam68 and PABPN1 in sensory ganglia neurons

    Neurotox Res

    (2010)
  • E.K. Mai et al.

    Phase III trial of bortezomib, cyclophosphamide and dexamethasone (VCD) versus bortezomib, doxorubicin and dexamethasone (PAd) in newly diagnosed myeloma

    Leukemia

    (2015)
  • R. Niesvizky et al.

    Community-based phase IIIB trial of three UPFRONT bortezomib-based myeloma regimens

    J Clin Oncol

    (2015)
  • P.G. Richardson et al.

    Reversibility of symptomatic peripheral neuropathy with bortezomib in the phase III APEX trial in relapsed multiple myeloma: impact of a dose-modification guideline

    Br J Haematol

    (2009)
  • M.A. Dimopoulos et al.

    Risk factors for, and reversibility of, peripheral neuropathy associated with bortezomib-melphalan-prednisone in newly diagnosed patients with multiple myeloma: subanalysis of the phase 3 VISTA study

    Eur J Haematol

    (2011)
  • S.K. Kumar et al.

    Impact of concomitant dexamethasone dosing schedule on bortezomib-induced peripheral neuropathy in multiple myeloma

    Br J Haematol

    (2017)
  • B. Hu et al.

    Efficacy and safety of subcutaneous versus intravenous bortezomib in multiple myeloma: a meta-analysis

    Int J Clin Pharmacol Ther

    (2017)
  • S.D. Mu et al.

    Subcutaneous versus intravenous bortezomib administration for multiple myeloma patients: a meta-analysis

    Curr Med Sci

    (2018)
  • Cited by (0)

    View full text