Elsevier

Leukemia Research

Volume 36, Supplement 1, November 2012, Pages S3-S12
Leukemia Research

Can we change the disease biology of multiple myeloma?

https://doi.org/10.1016/S0145-2126(12)70003-6Get rights and content

Abstract

Despite improvements in disease management, multiple myeloma (MM) remains incurable. Conventional treatment methods are unsatisfactory, leading to a pattern of regression and remission, and ultimately failure. This pattern suggests that one of the possible strategies for improving outcomes is continuous therapy to maintain suppression of the surviving tumor cells. Optimal management of MM requires potent agents and modalities with direct tumoricidal activity, which can also provide continuous suppression of the residual tumor to prevent disease relapse. Immunomodulatory agents exert immunomodulatory and tumoricidal effects, and cause disruption of stromal cell support from the bone marrow microenvironment. Therefore continuous therapy with immumomodulatory agents may be able to provide both tumor reduction and tumor suppression, enabling physicians to consider the possibility of incorporating continuous therapy into the treatment paradigm of patients with MM.

Introduction

Multiple myeloma (MM) is characterized by the accumulation of clonal plasma cells in the bone marrow, the presence of monoclonal immunoglobulin (Ig) in the serum or urine, osteolytic bone lesions, renal disease, and immunodeficiency. It is principally a disease of older patients, with a median age at diagnosis of 65–70 years. The first stage in the development of MM is the emergence of asymptomatic monoclonal gammopathy of undetermined significance (MGUS). In some of these patients, this progresses to smoldering MM and ultimately to symptomatic MM, with an annual risk of around 1% for patients with MGUS [1]. The reasons why MGUS progresses to MM in only a small proportion of patients are unclear, and both genetic and environmental factors have been implicated [2]. Progression to MM is associated with a series of complex genetic events in MM cells, as well as changes in the bone marrow microenvironment, including increased angiogenesis, suppression of the immune response, increased bone resorption, and the establishment of aberrant signalingloops involving cytokines and growth factors associated with the clinical features of MM and its resistance to treatment [3].

Despite the improvements in overall survival associated with the use of conventional high-dose chemotherapy and autologous stem-cell transplantation (HDT-ASCT), median overall survival remained at around 33 months until the introduction of the novel anti-myeloma agents, thalidomide, lenalidomide, and bortezomib [4]. For patients diagnosed since 2000, the use of the novel agents has improved survival times significantly, particularly for younger patients 5, 6. The median survival time for patients under 65 years of age treated with novel agents is 56 months [6]. The course of MM treated with conventional chemotherapy, such as single-agent alkylating drugs, corticosteroids or combination chemotherapy involving novel anti-myeloma agents is characterized by a pattern of remission and relapse, with a decreasing duration of response and increasing number of salvage regimens (Fig. 1). This reflects the development of drug resistance, which eventually results in refractory disease (Fig. 1) [7]. This pattern suggests the presence of residual disease after treatment, even following an apparently complete response. Therefore, the incurable nature of MM necessitates treatment with agents and modalities that not only provide direct tumoricidal effects to reduce tumor burden, but also suppress residual disease with continuous use.

The development of novel anti-MM agents relies on an understanding of the biology of MM and the multiple factors involved in its pathogenesis and response to treatment. As well as genetic aberrations in essential growth- and tumor-suppressor genes, there is increasing evidence that interactions between tumor cells and their bone marrow microenvironment play a pivotal role in the development, maintenance, and progression of MM, and thus, in the development of drug resistance. This knowledge has improved treatment options leading to the approval of drugs such as thalidomide, bortezomib, and lenalidomide, which not only target malignant cells directly, but also their supporting bone marrow microenvironment. In addition to their tumoricidal effects, immunomodulatory agents also act on the immune system, potentially helping to overcome MM-associated immunodeficiency and enhancing anti-MM immune activity.

This article aims to give an overview of the biology of MM, focusing on the pivotal role of the bone marrow microenvironment and its relevance to tumor survival and proliferation. It will also discuss how new agents have the potential to modify MM biology, offering the prospect of a shift in treatment paradigm to a focus on sustaining disease control with long-term treatment, which may transform myeloma into a chronic disease.

Section snippets

The biology of MM

The bone marrow of patients with MM contains malignant cells that have the morphology of mature plasma cells or plasmablasts. However, the origin of MM cells and their developmental relationship to non-malignant counterpart cells remains obscure. The vast majority of MM cells appear to be mature, quiescent, and terminally differentiated; therefore, they do not have long-term proliferative potential. This raises questions about which cells in MM patients are clonogenic and capable of

The role of the bone marrow microenvironment in MM pathogenesis

MM has become the prototypical tumor model for characterizing the interaction between tumor cells and their local milieu 34, 35. The bone marrow microenvironment refers not only to bone marrow stromal cells (BMSCs), but also to the non-cellular component composed of extracellular matrix (ECM) proteins such as collagen, fibronectin and laminin, and the extracellular fluid containing cytokines and growth factors. The bone marrow microenvironment supports normal hematopoiesis and these support

Immunodeficiency in MM

Immune dysfunction is an important feature of MM and is associated with an increased incidence of infections, which are a major cause of morbidity and mortality in myeloma patients. Importantly, immunodeficiency impacts disease progression and resistance to chemotherapy. Several factors produced as a result of MM cell-BMSC interactions also alter the functions of the host immune effector cells, thus interfering with immune surveillance and preventing immune-mediated tumor rejection [59].

MM

Mode of action of immunomodulatory agents

Thalidomide, a synthetic derivative of glutamic acid, has been found to have a range of properties including anti-inflammatory effects via inhibition of TNF-α [67], inhibition of angiogenesis [68], and immunomodulatory properties, including enhancement of T cell- and NK cell-mediated immunity 69, 70. These properties stimulated interest in thalidomide as an anticancer drug, particularly for the treatment of MM. Thalidomide has been relatively successful in improving survival in patients with

Direct tumoricidal effects and modulation of the tumor microenvironment

Immunomodulatory agents have been shown to have several direct and indirect effects on MM cells, via both direct tumoricidal effects and modulation of the bone marrow microenvironment, including the prevention of angiogenesis and osteoclastogenesis.

Lenalidomide down-regulates expression of the MM cell survival factor interferon regulatory factor-4 85, 86, 87. Conversely, it induces the expression of cyclin-dependent kinase inhibitors, including p21, p27, and p15, and the early response

Immunomodulatory properties

In vitro, immunomodulatory agents have been shown to augment both the adaptive and innate immune systems via enhancement of T-cell and NK-cell immune responses, both of which are reduced in MM patients 69, 89, 101, 102, 103. Immunomodulatory agents induce cell-surface expression of positive co-stimulatory molecules on T cells, including CD28, which is down-regulated in MM. T-cell co-stimulation by immunomodulatory agents via the B7-CD28 pathway is associated with up-regulation of cytokines,

Mode of action of proteasome inhibitors

Proteasome inhibitors act by targeting intracellular protein turnover via inhibition of the ubiquitin-proteasome pathway [113]. The proteasome inhibitor bortezomib, a specific inhibitor of the 26S proteasome, was the first of its class to enter clinical trials for the treatment of MM. Bortezomib is administered by injection and is indicated for the treatment of patients who have already received at least one prior therapy and undergone, or are unsuitable for, bone marrow transplantation, or

Direct tumoricidal effects and modulation of the tumor microenvironment

The therapeutic effects of bortezomib probably result from a combination of direct toxicity and its effects on the bone marrow microenvironment 3, 115. Proteasome inhibition results in cytoplasmic accumulation of IκB, which blocks the nuclear translocation and transcriptional activity of NF-κB. As discussed previously, inhibition of NF-κB results in a decrease in expression of a range of adhesion molecules and cytokines such as IL-6, which is involved in the growth and survival of MM cells. In

Effects on the immune system

Proteasome inhibition sensitizes tumor cells to NK cell-mediated lysis through TNF-related apoptosis-inducing ligand and/or Fas/Fas ligand pathways 126, 127. However, bortezomib has also been shown to disrupt NK-mediated immunity via induction of NK-cell apoptosis and suppression of NKp46 receptor-mediated cytotoxicity 128, 129. In addition, proteasome inhibitors induce apoptosis in activated and proliferating human T cells [130], interfere with the dendritic cell function [131] and antigen

Summary

In summary, MM is an incurable disease in which relapse is characterized by re-growth of residual tumor and immune suppression with a complex biology that affects many aspects of the disease and its response to treatment. As such, this disease requires effective long-term treatment strategies 135, 136. Conventional treatment, even with the addition of novel anti-myeloma agents, remains unsatisfactory. The disease is characterized by a pattern of regression and remission, and ultimately failure,

Conflict of interest statement

Dr. Borrello, MD, obtains research support and serves as a consultant to Celgene Corporation.

Acknowledgments

We thank Shanthi Jayawardena, PhD, and Eva Polk, PhD (Excerpta Medica), for writing assistance in the preparation of the manuscript. Editorial support was funded by Celgene Corporation. The authors were fully responsible for all content and editorial decisions for this manuscript.

References (136)

  • E Martinez-Garcia et al.

    The MMSET histone methyl transferase switches global histone methylation and alters gene expression in t(4; 14) multiple myeloma cells

    Blood

    (2011)
  • H Avet-Loiseau et al.

    Genetic abnormalities and survival in multiple myeloma: the experience of the Intergroupe Francophone du Myélome

    Blood

    (2007)
  • R Fonseca et al.

    Genomic abnormalities in monoclonal gammopathy of undetermined significance

    Blood

    (2002)
  • M Chesi et al.

    Frequent dysregulation of the c-maf proto-oncogene at 16q23 by translocation to an Ig locus in multiple myeloma

    Blood

    (1998)
  • R Fonseca et al.

    Clinical and biologic implications of recurrent genomic aberrations in myeloma

    Blood

    (2003)
  • CS Mitsiades et al.

    The role of the bone microenvironment in the pathophysiology and therapeutic management of multiple myeloma: interplay of growth factors, their receptors and stromal interactions

    Eur J Cancer

    (2006)
  • CS Mitsiades et al.

    The role of the bone marrow microenvironment in the pathophysiology of myeloma and its significance in the development of more effective therapies

    Hematol Oncol Clin North Am

    (2007)
  • G Teoh et al.

    Interaction of tumor and host cells with adhesion and extracellular matrix molecules in the development of multiple myeloma

    Hematol Oncol Clin North Am

    (1997)
  • D Chauhan et al.

    Multiple myeloma cell adhesion-induced interleukin-6 expression in bone marrow stromal cells involves activation of NF-kappa B

    Blood

    (1996)
  • H Uchiyama et al.

    Adhesion of human myeloma-derived cell lines to bone marrow stromal cells stimulates interleukin-6 secretion

    Blood

    (1993)
  • T Hideshima et al.

    Targeting p38 MAPK inhibits multiple myeloma cell growth in the bone marrow milieu

    Blood

    (2003)
  • B Dankbar et al.

    Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma

    Blood

    (2000)
  • D Chauhan et al.

    Interleukin-6 inhibits Fas-induced apoptosis and stress-activated protein kinase activation in multiple myeloma cells

    Blood

    (1997)
  • CW Löwik et al.

    Parathyroid hormone (PTH) and PTH-like protein (PLP) stimulate interleukin-6 production by osteogenic cells: a possible role of interleukin-6 in osteoclastogenesis

    Biochem Biophys Res Commun

    (1989)
  • J Moreaux et al.

    BAFF and APRIL protect myeloma cells from apoptosis induced by interleukin 6 deprivation and dexamethasone

    Blood

    (2004)
  • P Georgii-Hemming et al.

    Insulin-like growth factor I is a growth and survival factor in human multiple myeloma cell lines

    Blood

    (1996)
  • CS Mitsiades et al.

    Inhibition of the insulin-like growth factor receptor-1 tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematologic malignancies, and solid tumors

    Cancer Cell

    (2004)
  • RH Prabhala et al.

    Elevated IL-17 produced by Th17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma

    Blood

    (2010)
  • K Noonan et al.

    A novel role of IL-17-producing lymphocytes in mediating lytic bone disease in multiple myeloma

    Blood

    (2010)
  • G Cook et al.

    Immune regulation in multiple myeloma: the host-tumour conflict

    Blood Rev

    (1999)
  • M Ratta et al.

    Dendritic cells are functionally defective in multiple myeloma: the role of interleukin-6

    Blood

    (2002)
  • M Urashima et al.

    Transforming growth factor-beta1: differential effects on multiple myeloma versus normal B cells

    Blood

    (1996)
  • FE Davies et al.

    Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma

    Blood

    (2001)
  • R LeBlanc et al.

    Immunomodulatory drug costimulates T cells via the B7-CD28 pathway

    Blood

    (2004)
  • MQ Lacy et al.

    Pomalidomide plus low-dose dexamethasone in myeloma refractory to both bortezomib and lenalidomide: comparison of 2 dosing strategies in dual-refractory disease

    Blood

    (2011)
  • YX Zhu et al.

    Cereblon expression is required for the antimyeloma activity of lenalidomide and pomalidomide

    Blood

    (2011)
  • S Li et al.

    IMiD immunomodulatory compounds block C/EBPβ translation through eIF4E down-regulation resulting in inhibition of MM

    Blood

    (2011)
  • N Mitsiades et al.

    Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: therapeutic implications

    Blood

    (2002)
  • T Hideshima et al.

    Thalidomide and its analogs overcome drug resistance of human multiple myeloma cells to conventional therapy

    Blood

    (2000)
  • K Dredge et al.

    Orally administered lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endothelial cell migration and Akt phosphorylation in vitro

    Microvasc Res

    (2005)
  • J Jakubikova et al.

    Lenalidomide targets clonogenic side population in multiple myeloma: pathophysiologic and clinical implications

    Blood

    (2011)
  • RA Kyle et al.

    A long-term study of prognosis in monoclonal gammopathy of undetermined significance

    N Engl J Med

    (2002)
  • RA Kyle et al.

    The significance of monoclonal gammopathy of undetermined significance

    Haematologica

    (2009)
  • RA Kyle et al.

    Multiple myeloma

    N Engl J Med

    (2004)
  • I Turesson et al.

    Patterns of improved survival in patients with multiple myeloma in the twenty-first century: a population-based study

    J Clin Oncol

    (2010)
  • CA Huff et al.

    Multiple myeloma cancer stem cells

    J Clin Oncol

    (2008)
  • R Bataille et al.

    The phenotype of normal, reactive and malignant plasma cells. Identification of “many and multiple myelomas” and of new targets for myeloma therapy

    Haematologica

    (2006)
  • J Cao et al.

    A CD10-positive subset of malignant cells is identified in multiple myeloma using PCR with patient-specific immunoglobulin gene primers

    Leukemia

    (1995)
  • K Kiel et al.

    Analysis of circulating tumor cells in patients with multiple myeloma during the course of high-dose therapy with peripheral blood stem cell transplantation

    Bone Marrow Transplant

    (1999)
  • H Kubagawa et al.

    Studies on the clonal origin of multiple myeloma. Use of individually specific (idiotype) antibodies to trace the oncogenic event to its earliest point of expression in B-cell differentiation

    J Exp Med

    (1979)
  • Cited by (48)

    • Zinc oxide nanoparticles induce human multiple myeloma cell death via reactive oxygen species and Cyt-C/Apaf-1/Caspase-9/Caspase-3 signaling pathway in vitro

      2020, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      It is characterized by a large number of plasma cells in bone marrow [1]. The main clinical manifestations of MM patients are anemia, renal insufficiency, immunodeficiency, infection and hypercalcemia [2,3]. MM accounts for 1 % of all cancer cases and 13 % of hematological neoplasms [4].

    • Lenalidomide and Vorinostat Maintenance after Autologous Transplantation in Multiple Myeloma: Long- Term Follow-Up

      2020, Biology of Blood and Marrow Transplantation
      Citation Excerpt :

      However, ASCT is not curative, and most patients experience progressive disease (PD), even in those who attain a complete response (CR). Maintenance therapy has been used as a viable treatment strategy for control or elimination of residual disease [4-6]. Several strategies have been assessed in this setting, with lenalidomide, an immunomodulatory drug, showing the most promise because of its mode of action and favorable toxicity profile [7,8].

    • The emerging roles and clinical potential of exosomes in cancer: Drug resistance

      2018, Diagnostic and Therapeutic Applications of Exosomes in Cancer
    • Bone marrow B lymphocytes in multiple myeloma and MGUS: Focus on distribution of naïve cells and memory subsets

      2016, Leukemia Research
      Citation Excerpt :

      Multiple myeloma (MM) is a disease principally of older patients, with a median age at diagnosis of 65–70 years [1]. It is caused by proliferation of clonal plasma cells (cPCs) in the bone marrow (BM), associated with the presence of monoclonal immunoglobulin (M component) in the serum or urine, and signs of multi-organ impairment that may include anemia, lytic bone lesions, immunodeficiency, and reduced renal function [1–3]. MM usually evolves from an asymptomatic premalignant stage called monoclonal gammopathy of undetermined significance (MGUS), which is associated with a risk of progression to myeloma of 1% per year [1–3].

    View all citing articles on Scopus
    View full text