A New and Evolving Era in Multiple Myeloma Field

Special Article - Multiple Myeloma

Ann Hematol Oncol. 2016; 3(6): 1097.

A New and Evolving Era in Multiple Myeloma Field

Ni J*

Department of Hematology/Oncology Pharmacy, Brigham and Women’s Hospital, USA

*Corresponding author: Jian Ni, Department of Hematology/Oncology Pharmacy, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, USA

Received: June 21, 2016; Accepted: August 03, 2016; Published: August 05, 2016

Abstract

Multiple myeloma (MM) is one of the most common hematologic disorders in US, which accounts for approximately 15% of hematologic malignancies [1]. The American Cancer Society estimates that about 30,330 people will be diagnosed with multiple myeloma and about 12,650 will die of the disease in 2016 [1]. The number of people diagnosed with MM has been increasing over the past ten years, fortunately, with the deeper understanding of the disease and advances in treatment and research, more novel therapeutic classes have been granted FDA approval and incorporated into treatment leading to treatment paradigm change and significantly improved patients’ survival [2]. To address recent discoveries about the molecular pathogenesis of the disease, International myeloma working group (IMWG) has updated diagnostic criteria and staging system for Multiple myeloma [3,4]. Recently National Comprehensive Cancer Network (NCCN) has updated its guideline to reflect rapid progress in the management of multiple myeloma [5]. The purpose of this review is to highlight some of most recent changes and significant progress in multiple myeloma clinical practice from basic disease definition changes to the advancement of therapeutic options including most recently FDA approved anti-myeloma agents as well as some promising novel therapeutic approaches under clinical development.

Keywords: Multiple myeloma (MM); Revised international staging system (R-ISS); Minimal residual disease (MRD); Risk stratification; Novel therapeutics; Immunotherapy

Abbreviations

MM: Multiple Myeloma; MRD: Minimal Residual Disease; CR: Complete Response; OS: Overall Survival; PFS: Progression Free Survival; TTP: Time To Progression; NGS: Next-Generation Sequencing; CDC: Complement Dependent Cytotoxicity; ADCC: Antibody-Dependent Cellular Cytotoxicity; BCMA: B-cell Maturation Antigen; CRS: Cytokine Release Syndrome; FISH: Fluorescence In Situ Hybridization; R-ISS: Revised International Staging System; PD-1: Programmed T cell death 1; PD-L1: Programmed Death- Ligand1; HDT/ASCT: High-Dose Therapy/Autologous Stem Cell Transplantation; NCCN: National Comprehensive Cancer Network; IMWG: International Myeloma Working Group; MFC: Multiparameter-Flow-Cytometry; CRd: Carfilzomib Combined with Lenalidomid and Dexamethasone; ERd : Elotuzumab in combination with Lenalidomide and Dexamethasone; RRMM: Relapsed or Refractory Multiple Myeloma; NDMM: Newly Diagnosed Multiple Myeloma; CAR: Chimeric Antigen Receptor T cells; PET/CT: Positron Emission Tomography Computed Tomography; LDH: Lactate Dehydrogenase; SMM: Smoldering Multiple Myeloma

Introduction

Multiple myeloma (MM) is one of the most common hematologic disorders in US, which accounts for approximately 15% of hematologic malignancies [1]. The American Cancer Society estimates that more than 30,000 people will be diagnosed with multiple myeloma and nearly 13,000 will die of the disease this year [1]. The number of people diagnosed with MM has been increasing over the past ten years, fortunately, with the deeper understanding of the disease and advances in treatment and research, more novel therapeutic classes have been granted FDA approval and incorporated into treatment, which has significantly prolonged patients’ survival [2]. To address recent discoveries about the molecular pathogenesis of the disease, International myeloma working group (IMWG) has redefined the basic diagnosis and response criteria [3,4]. Recently National Comprehensive Cancer Network (NCCN) has updated its guideline to reflect rapid progress in the management of multiple myeloma [5]. The purpose of this review is to highlight some of most recent changes and significant progress in multiple myeloma clinical practice from basic disease definitions to the advancement of therapeutic options.

Basic disease definitions and criteria

Revised diagnostic criteria: Historically, when the therapeutic options were limited and there was no apparent benefit from early treatment, multiple myeloma (MM) treatment would not be initiated if there was no sign of end-organ damage described in CRAB criteria. However, with emergence with novel agents and advanced technologies, the 5 year survival rate has been greatly improved and prevention of end-organ damage has become important [3]. To address this issue, the international myeloma Working Group (IMWG) updated definition of MM to include biomarkers in addition to existing requirements of CRAB features [4]. The diagnosis requires 10% or more clonal plasma cells on bone marrow examination or a biopsy proven plasmacytoma plus evidence of associated end-organ damage. Even if end-organ damage is not present, patients with = 60% bone marrow plasmacytosis, involved: uninvolved serum free lightchain ratio =100 with the involved serum free light-chain =10 mg/dL, or more than 1 focal lesion on magnetic resonance imaging (MRI) studies are eligible for treatment [4]. The New IMWG diagnostic criteria not only allow for early therapy which could potentially improve survival, but also incorporate technology advances encouraging the use of modern imaging methods to make an early diagnosis. This new diagnostic criteria is included in the recently published National Comprehensive Cancer Network (NCCN) 2016 version of the Clinical Practice Guidelines in Oncology for Multiple Myeloma (MM) [5].

Updated response criteria: Based on Data from multiple clinical trials, the achievement of Complete Response (CR) has been shown to be a prognostic indicator of long-term progression-free survival (PFS) and Overall survival (OS) in patients treated by HDT/ASCT and novel agents, regardless of age, ISS stage and treatment [6]. Conventional Complete Response (CR), defined as disappearance of M-protein on immunofixation, on survival of MM has generated controversy in current literatures and in clinical practice. Improvements in techniques for assessing disease status have led to development of more stringent response criteria. As a result, IMWG further categorizes complete response into molecular CR, immunophenotypic CR and astringent CR [4]. With the new criteria, assessment of treatment response is transitioning from conventional CR to MRD negative status assessment and Stringent Complete Response in MM patients.

Minimal Residual Disease (MRD) refers to the persistence of small number of residual myeloma cells during or following treatment. Presence of MRD that is below the sensitivity of bone marrow (BM) morphology, protein electrophoresis with immunofixation and light chain quantitation will eventually lead to relapse, even in patient with CR [7]. Developments in new diagnostic techniques have allowed for the detection of minimal residual disease, which has emerged as one of the most relevant prognostic factors in both transplant candidates and elderly patients who are not considered to be transplant-eligible, irrespectively of patients’ age and cytogenetic risk [8]. Response criteria studies have shown that among patients achieving a biochemical CR, MRD-negative status is associated with superior outcomes including progression-free survival (PFS) and OS [8-10]. In a multicenter randomized phase 3 trial, each log depletion of MRD was associated with significant improvement of OS (median OS of 1, 4, 5.9, 6.8 and >7.5 years for MRD =10%, < 10%, 0.1% to < 1%, 0.01% to < 0.1% and < 0.01% respectively) [8].

In order to sufficiently assess the deepness of response, more sensitive method should be utilized to measure and monitor MRD. Research shows that persistent MRD detected by multiparameterflow- cytometry (MFC), polymerase-chain-reaction (PCR), Nextgeneration sequencing (NGS) indicates poor clinical outcome among patients with complete remission [11]. One recent study shows that using 2nd generation MFC, immune profiling concomitant to MRD monitoring also contributed to identify patients with different outcomes, being poor, intermediate and favorable outcome (25%, 61% and 100% OS at 3-years; P=.01) in the study) [7]. Newer imaging modalities including PET/CT and MRI scans have also been included in disease assessment and progression criteria [8].

Rawstron and colleagues report that a lower cutoff of 0.01% threshold (10−4) using more sensitive assays (e.g., next generation sequencing (NGS) or high-sensitive multiparameter flow cytometry (MFC)) will likely provide a better assessment of clinical outcome [8]. Martinez-Lopez’s study further identified 3 groups of patients with different time to progression (TTP) using NGS: patients with high (<10−3), intermediate (10−3 to 10−5) and low (>10−5) MRD levels showed significantly different TTP (27, 48 and 80 months, respectively). According to this study, 10−5 could be considered as the target cutoff level for definition of MRD negativity [9]. MRD monitoring can be used to evaluate the efficacy of different treatment strategies and tailored therapy in MM. It is predictable that there will be increased reliance on minimal residual disease testing to guide therapy. Future efforts need to address systematic usage of highly sensitive, cost-effective and standardized MRD assessment techniques.

Cytogenetics and risk stratification: MM is characterized by significant tumor heterogeneity with clonal evolution affecting both prognostic stratification and therapeutic approaches. Genome instability represents an important feature observed in MM cells leading to emergence of genetic change which ultimately results in acquisition of drug resistance and disease progression [12]. Moreover, bone marrow microenvironment plays a role in supporting MM cell survival, proliferation and drug resistance [13]. Comprehensive oncogenomic analysis has identified many complex genetic and epigenetic alterations in multiple myeloma. A number of genetic aberrations such as t(4; 14) and 17p13 deletion have been shown to be associated with poor survival consistently across studies [14], whereas others are controversial with conflicting evidence in the literature. Although FISH-identified t(4;14), t(14;16) and del17p13 have been considered to convey poor prognosis [15], more recent single-nucleotide polymorphism array analysis in 192 uniformly treated patients identified amp(1q23.3), amp(5q31.3) and del(12p13.31) as the most powerful independent adverse prognostic markers (P< 0.0001) [15]. Even within groups with these genetic prognostic factors, there may be further heterogeneity. For example, the IFM group showed that among the patients with t(4; 14), those with a hemoglobin greater than 10 g/l and beta-2 microglobulin less than 4 mg/l had significantly longer survival compared to those without [15]. At the same time, a recent analysis showed that patients with high-risk genetic changes have significantly different survival depending on the presence or absence of trisomies [16]. Based on these findings, genetics alone should not be considered as prognostic factors. Combining information about genetic abnormalities with other parameters may improve their prognostic value. Future study could focus on clinical translation of acquired epigenetic knowledge such as genome instability and the exploitable vulnerabilities it creates, which could eventually lead to development of new treatment options [12].

The consensus statement on risk stratification released by the International Myeloma Working Group (IMWG) combined both disease burden and biology features for patients with multiple myeloma. The new IMWG risk stratification has three risk categories: low-risk, standard-risk and high-risk. A patient’s risk classification is based on the disease stage according to the International Staging System (ISS), the presence of certain chromosomal abnormalities in the patient’s myeloma cells based on results of FISH testing and patient’s age [14]. Patients who are ISS stage II or III and whose myeloma cells contain the translocation t(4; 14) or the deletion del(17p13) are classified as high-risk [14]. IMWG advocates use of ISS staging in conjunction with FISH for t(4; 14), deletion 17p13 and 1q21 gain for risk stratification in MM [14] (Table 1).

Citation: Ni J. A New and Evolving Era in Multiple Myeloma Field. Ann Hematol Oncol. 2016; 3(6): 1097. ISSN : 2375-7965