Chapter 3 - Functional and Clinical Relevance of Chondroitin Sulfate Proteoglycan 4

https://doi.org/10.1016/B978-0-12-380890-5.00003-XGet rights and content

Abstract

The lack of effective conventional therapies for the treatment of advanced stage melanoma has stimulated interest in the development of novel strategies for the management of patients with malignant melanoma. Among them, immunotherapy has attracted much attention because of the potential role played by immunological events in the clinical course of melanoma. For many years, T cell-based immunotherapy has been emphasized in part because of the disappointing results of the monoclonal antibody (mAb)-based clinical trials conducted in the early 1980s and in part because of the postulated major role played by T cells in tumor growth control. More recently, mAb-based therapies have gained in popularity given their clinical and commercial success for a variety of malignant diseases. As a result, there has been increased interest in identifying and characterizing antibody-defined melanoma antigens. Among them, the chondroitin sulfate proteoglycan 4 (CSPG4), also known as high molecular weight-melanoma associated antigen (HMW-MAA) or melanoma chondroitin sulfate proteoglycan (MCSP), has attracted much attention in recent years because of the growing experimental evidence that it fulfills two requirements for immunotherapy to be therapeutically effective: (1) targeting of cancer stem cells (CSC) and (2) development of combinatorial therapies to counteract the escape mechanisms driven by the genetic instability of tumor cells. With this in mind, in this chapter, we have reviewed recent information related to the distribution of CSPG4 on various types of tumors, including CSC, its expression on pericytes in the tumor microenvironment, its recognition by T cells, its role in cell biology as well as the potential mechanisms underlying the ability of CSPG4-specific immunity to control malignant cell growth.

Introduction

Malignant melanoma represents the most common form of fatal skin cancer. Current estimates indicate that its incidence is increasing at a rate of 5% per year (MacKie et al., 2009). In spite of significant improvements in diagnosis (Terando et al., 2003), the mortality rate for melanoma has been continually increasing over the past decade and the disease represents one of the most common fatal malignancies of young adults (MacKie et al., 2009). While early stage melanoma is highly curable with surgery (Crosby et al., 2001), advanced stage melanoma is relatively resistant to conventional therapeutic regimens and is often fatal (Cascinelli et al., 2003, Cooper, 2002, Soengas & Lowe, 2003). Although promising results have been obtained in recent clinical trials with inhibitors of the extracellular signal-related MAP kinase pathway, such as B-Raf and MEK inhibitors (Flaherty et al., 2010), the lack of effective conventional therapies for the treatment of advanced stage disease has stimulated interest in the application of novel strategies for the management of patients with malignant melanoma. Among them, immunotherapy has been emphasized in recent years for a number of reasons. First, the immune system appears to play a role in the natural history of melanoma (Guerry, 1998). Second, the recent success of monoclonal antibody (mAb)-based therapies in patients with some hematological malignancies and some solid tumors (Campoli and Ferrone, 2009), the Federal Drug Administration (FDA) approval of interferon-α2b (Kirkwood et al., 1996) and interleukin-2 (IL-2) (Atkins et al., 1999) for use as adjuvant therapy for patients with advanced stage melanoma as well as the promising results obtained with the CTLA-4 specific mAb ipilimumab in patients with melanoma (Hodi et al., 2010) and the recent FDA approval of PROVENGE® (sipuleucel-T), an autologous cellular vaccine designed to stimulate T-cell immunity to prostatic acid phosphatase in patients with asymptomatic or minimally symptomatic metastatic (hormone refractory) prostate cancer (Cha and Fong, 2010), provide a rationale for the use of immunotherapy to treat patients with melanoma. Third, the identification of well-characterized melanoma antigens (MA) (Ribas et al., 2003, Stevanovic, 2002) and the development of their corresponding probes, that is, mAb (Weiner et al., 2010) and cytotoxic T lymphocytes (CTL) (Rosenberg and Dudley, 2009), have provided moieties to target melanoma lesions with highly specific reagents. Fourth, the substantial increase in our understanding of the molecular events leading to an immune response as well as the development of effective immunization strategies have facilitated the application of immunotherapy for the treatment of melanoma (Alexandrescu et al., 2010).

To date, a number of MA that meet the criteria to be utilized for immunotherapy, that is, high expression in a large percentage of melanoma lesions and restricted distribution in normal tissues, have been identified and utilized to implement clinical trials in patients with melanoma. During the past 20 years, the use of T cell-defined MA has been emphasized because of the disappointing results obtained with mAb-based immunotherapy in the early 1980s (Milstein and Waldmann, 1999) and because of the general belief that T cells play a major role in tumor growth control (Dudley et al., 2002, Yee et al., 2002). More recently, mAb-based therapies have gained in popularity given their clinical and commercial success for a variety of malignant diseases (Campoli and Ferrone, 2009). The beneficial clinical effects of mAb-based immunotherapy are believed to reflect the ability of antigen-specific mAb to (i) inhibit tumor cell proliferation; (ii) induce tumor cell apoptosis; (iii) trigger antibody-dependent cell-mediated cytotoxicity (ADCC); (iv) mediate complement-dependent cytotoxicity (CDC); and (v) interfere with the function of the targeted antigen and/or affect tumor cell signaling (Campoli and Ferrone, 2009). More recent evidence suggests that tumor antigen (TA)-specific mAb can induce TA-specific T cell immune responses (Campoli et al., 2010). We have focused our studies on antibody-defined MA, since mAb can utilize both immunological as well as nonimmunological mechanisms to control tumor growth. As a result, they are less susceptible to the escape mechanisms caused by defects in TA presentation which are frequent in melanoma cells (Marincola et al., 2000).

The MA that has been the focus of our studies over the past 30 years is the chondroitin sulfate proteoglycan 4 (CSPG4), also known as high molecular weight-melanoma associated antigen (HMW-MAA or melanoma chondroitin sulfate proteoglycan (MCSP) (Campoli et al., 2004). CSPG4 represents an attractive target, since it has a restricted distribution in normal tissues and is expressed in a large percentage of melanoma lesions, on putative cancer stem cells (CSC) and on activated pericytes in the tumor microenvironment. Furthermore, CSPG4 is recognized by both antibodies and T cells. As a result, CSPG4 provides the opportunity to apply combinatorial antibody- and T cell-based immunotherapy to target not only tumor cells including CSC, but also cells in the tumor microenvironment that are required for tumor cell survival and growth.

A considerable amount of information has been accumulated in recent years regarding (1) the distribution and expression of CSPG4 on malignant cells as well as on both normal stem cells and CSC; (2) the role CSPG4 plays in the biology of both normal and malignant cells, and (3) the ability of the adaptive immune system to target CSPG4. In this review, we highlight the properties of CSPG4, that make it a suitable target of immunotherapy for the treatment of patients with malignant disease, mainly emphasizing the information published during the past 5 years and the results of studies in progress in our laboratory. However, we have revisited several concepts regarding CSPG4 that have been derived from previous investigations, since they now need to be reinterpreted in light of the results which have recently become available. Additionally, we have discussed the role CSPG4 plays in the biology of malignant cells with emphasis on the recently identified signal transduction pathways triggered by CSPG4. Finally, we have discussed the potential mechanisms underlying the ability of CSPG4-specific immunity to control malignant cell growth.

Section snippets

Phylogenetic Evolution, Structure, and Tissue Distribution of CSPG4

In the early 1980s the interest in the use of CSPG4 as a target of immunotherapy for the treatment of melanoma and the availability of mAb provided the stimulus to investigate CSPG4 expression in melanoma lesions and in normal tissues, the structural profile of CSPG4 as well as the cross-reactivity of human CSPG4-specific mAb with xenogeneic homologues of human CSPG4. More recently taking advantage of the information about the nucleotide sequence of xenogeneic homologues of CSPG4, its

Functional Properties of CSPG4

Early investigations had shown that NG2, the rat homologue of CSPG4, promotes progenitor and tumor cell motility, adhesion, and growth, resulting in melanoma and glioma cell growth and metastasis (Stallcup and Huang, 2008). More recently, in vitro and in vivo assays with CSPG4-specific mAb have shown that the functional properties of CSPG4 are similar to those displayed by NG2.

Clinical Applications of CSPG4

The expression of CSPG4 in a high percentage of melanoma lesions with low intra- and interlesional heterogeneity, its restricted distribution in normal tissues, and the availability of CSPG4-specific mAb have provided the rationale to utilize CSPG4 as a marker for immunoscintigraphy of malignant disease lesions and as a target of immunotherapy.

Conclusion

In the past 10 years, there has been significant progress in the characterization of the expression of CSPG4 by normal and malignant cells, its role in the biology of melanoma cells and its recognition by humoral and cellular immunity. The information we have acquired has greatly contributed to our understanding of the unique features of this antigen and of its potential as a target of immunotherapy. Nonetheless, a number of questions remain; we address some of them in this section of this

Acknowledgments

This work was supported by The Empire Grant NYSDOH C017927, The Elsa U. Pardee Foundation (X.W.), and by the PHS grants R01 CA105500, R01 CA110249, and R01 CA138188 (S.F.), awarded by the National Cancer Institute.

References (188)

  • R.O. Dillman

    Treatment of low-grade B-cell lymphoma with the monoclonal antibody rituximab

    Semin. Oncol.

    (2003)
  • S. Ferrone et al.

    Human high molecular weight-melanoma associated antigen mimicry by mouse anti-idiotypic monoclonal antibodies MK2-23. Experimental studies and clinical trials in patients with malignant melanoma

    Pharmacol. Ther.

    (1993)
  • R. Forstpointner et al.

    The addition of rituximab to a combination of fludarabine, cyclophosphamide, mitoxantrone (FCM) significantly increases the response rate and prolongs survival as compared with FCM alone in patients with relapsed and refractory follicular and mantle cell lymphomas: Results of a prospective randomized study of the German Low-Grade Lymphoma Study Group

    Blood

    (2004)
  • N.Y. Frank et al.

    Regulation of progenitor cell fusion by ABCB5 P-glycoprotein, a novel human ATP-binding cassette transporter

    J. Biol. Chem.

    (2003)
  • L. Ghali et al.

    Epidermal and hair follicle progenitor cells express melanoma-associated chondroitin sulfate proteoglycan core protein

    J. Invest. Dermatol.

    (2004)
  • L. Goretzki et al.

    High-affinity binding of basic fibroblast growth factor and platelet-derived growth factor-AA to the core protein of the NG2 proteoglycan

    J. Biol. Chem.

    (1999)
  • K.A. Grako et al.

    Participation of the NG2 proteoglycan in rat aortic smooth muscle cell responses to platelet-derived growth factor

    Exp. Cell Res.

    (1995)
  • S.I. Grivennikov et al.

    Dangerous liaisons: STAT3 and NF-kappaB collaboration and crosstalk in cancer

    Cytokine Growth Factor Rev.

    (2010)
  • J.M. Hilden et al.

    MLL gene rearrangement, cytogenetic 11q23 abnormalities, and expression of the NG2 molecule in infant acute myeloid leukemia

    Blood

    (1997)
  • J. Iida et al.

    Cell surface chondroitin sulfate proteoglycans in tumor cell adhesion, motility and invasion

    Semin. Cancer Biol.

    (1996)
  • J. Iida et al.

    A role of chondroitin sulfate glycosaminoglycan binding site in alpha4beta1 integrin-mediated melanoma cell adhesion

    J. Biol. Chem.

    (1998)
  • J. Iida et al.

    Melanoma chondroitin sulfate proteoglycan regulates matrix metalloproteinase-dependent human melanoma invasion into type I collagen

    J. Biol. Chem.

    (2001)
  • K. Imai et al.

    Higher cytolytic efficiency of an IgG2 alpha than of an IgG1 monoclonal antibody reacting with the same (or spatially close) determinant on a human high-molecular-weight melanoma-associated antigen

    Cell. Immunol.

    (1982)
  • W.M. Klein et al.

    Increased expression of stem cell markers in malignant melanoma

    Mod. Pathol.

    (2007)
  • I. Kozanoglu et al.

    Human bone marrow mesenchymal cells express NG2: Possible increase in discriminative ability of flow cytometry during mesenchymal stromal cell identification

    Cytotherapy

    (2009)
  • G. Kraal et al.

    High endothelial venules: Lymphocyte traffic control and controlled traffic

    Adv. Immunol.

    (1997)
  • T.S. Lin et al.

    Rituximab in B-cell chronic lymphocytic leukemia

    Semin. Oncol.

    (2003)
  • D.T. Alexandrescu et al.

    Immunotherapy for melanoma: Current status and perspectives

    J. Immunother.

    (2010)
  • M.B. Atkins et al.

    High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: Analysis of 270 patients treated between 1985 and 1993

    J. Clin. Oncol.

    (1999)
  • D.S. Barritt et al.

    The multi-PDZ domain protein MUPP1 is a cytoplasmic ligand for the membrane-spanning proteoglycan NG2

    J. Cell. Biochem.

    (2000)
  • S. Basak et al.

    Induction of cellular immunity by anti-idiotypic antibodies mimicking GD2 ganglioside

    Cancer Immunol. Immunother.

    (2003)
  • M.S. Benassi et al.

    NG2 expression predicts the metastasis formation in soft-tissue sarcoma patients

    J. Orthop. Res.

    (2009)
  • H. Bender et al.

    Effects of diagnostic application of monoclonal antibody on survival in melanoma patients

    Hybridoma

    (1997)
  • M. Bernfield et al.

    Functions of cell surface heparan sulfate proteoglycans

    Annu. Rev. Biochem.

    (1999)
  • C. Bluemel et al.

    Epitope distance to the target cell membrane and antigen size determine the potency of T cell-mediated lysis by BiTE antibodies specific for a large melanoma surface antigen

    Cancer Immunol. Immunother.

    (2010)
  • A.D. Boiko et al.

    Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271

    Nature

    (2010)
  • G.M. Bokoch

    Biology of the p21-activated kinases

    Annu. Rev. Biochem.

    (2003)
  • J. Bomanji et al.

    Characterisation of ocular melanoma with cutaneous melanoma antibodies

    Br. J. Ophthalmol.

    (1987)
  • A. Borkhardt et al.

    Infant acute lymphoblastic leukemia—Combined cytogenetic, immunophenotypical and molecular analysis of 77 cases

    Leukemia

    (2002)
  • E.E. Bosco et al.

    Rac1 GTPase: A “Rac” of all trades

    Cell. Mol. Life Sci.

    (2009)
  • M.A. Burg et al.

    Expression of the NG2 proteoglycan enhances the growth and metastatic properties of melanoma cells

    J. Cell. Physiol.

    (1998)
  • M. Campoli et al.

    Immunotherapy of malignant disease: The coming age of therapeutic monoclonal antibodies

  • M. Campoli et al.

    Human high molecular weight-melanoma-associated antigen (HMW-MAA): A melanoma cell surface chondroitin sulfate proteoglycan (MSCP) with biological and clinical significance

    Crit. Rev. Immunol.

    (2004)
  • M. Campoli et al.

    Immunotherapy of malignant disease with tumor antigen-specific monoclonal antibodies

    Clin. Cancer Res.

    (2010)
  • E. Cha et al.

    Therapeutic vaccines for prostate cancer

    Curr. Opin. Mol. Ther.

    (2010)
  • J.C. Challier et al.

    Characterization of first trimester human fetal placental vessels using immunocytochemical markers

    Cell. Mol. Biol. (Noisy-le-grand)

    (2001)
  • P. Chattopadhyay et al.

    Murine monoclonal anti-idiotope antibody breaks unresponsiveness and induces a specific antibody response to human melanoma-associated proteoglycan antigen in cynomolgus monkeys

    Proc. Natl. Acad. Sci. USA

    (1992)
  • M. Chekenya et al.

    NG2 proteoglycan promotes angiogenesis-dependent tumor growth in CNS by sequestering angiostatin

    FASEB J.

    (2002)
  • G. Clauss et al.

    Melanoma associated antigens in Xiphophorus Fish

  • B.D. Cox et al.

    New concepts regarding focal adhesion kinase promotion of cell migration and proliferation

    J. Cell. Biochem.

    (2006)
  • Cited by (96)

    • Skin cancer biology and its biomarkers: Recent trends and prospective

      2023, Biomarkers in Cancer Detection and Monitoring of Therapeutics: Diagnostic and Therapeutic Applications: Volume 2
    • CAR T-cell therapy for triple-negative breast cancer: Where we are

      2020, Cancer Letters
      Citation Excerpt :

      Therefore, TEM8 expression should be thoroughly examined in healthy human tissues, and these findings warrant careful consideration as L2-based TEM8 CART therapeutic approaches move into the clinical trials. CSPG4, also known as melanoma-associated chondroitin sulfate proteoglycan (MCSP) or high molecular weight melanoma-associated antigen (HMW-MAA), is a type 1 transmembrane proteoglycan that plays an important role in growth, motility, and survival of melanoma cells [57]. CSPG4 displays a broad normal tissue distribution, such as lung, heart, central nervous system, lymphoid organs, intestinal tissue skin, smooth muscle, pericytes, adipose tissue, uterus, and prostate at the RNA level [58].

    • Role of cell surface proteoglycans in cancer immunotherapy

      2020, Seminars in Cancer Biology
      Citation Excerpt :

      Also, CAR-NK-GPC-3 therapy resulted in decreased tumor proliferation, increased tumor apoptosis and the expression of GPC-3 and TGF-β (the main molecule associated with the induction of EMT) [205]. Chondroitin sulphate proteoglycan-4 (CSPG-4) is another cell surface PG which is aberrantly expressed in cancer and which is associated with the malignant behavior of various types of cancer including melanoma, breast cancer, squamous cell carcinoma of the head and neck (SCCHN), basal breast cancer, mesothelioma, pancreatic carcinoma, some types of renal cell carcinoma, and hematopoietic malignancies [206]. In malignant mesothelioma, the CSPG4-specific mAb TP41.2 inhibited the growth of human xenografts in immunodeficient mice, and significantly extended animal survival [207].

    View all citing articles on Scopus
    View full text