LYMPHOID NEOPLASIA Dendritic cells accumulate in the bone marrow of myeloma patients where they protect tumor plasma cells from CD8 1 T-cell killing

Many researchers have speculated that the clinical progression from monoclonal gammop-athy of undetermined significance (MGUS) to multiple myeloma (MM) is driven by defects in dendritic cell (DC) function. However, evidence supporting this assumption is controversial, and no mechanism for the putative DC dysfunction has been demonstrated thus far. We studied DC subsets from the bone marrow of MM patients compared with those of MGUS patients and control subjects. We found that myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) accumulate in the bone marrow during the MGUS-to-MM progression. After engulfmentofapoptotictumorplasmacellsviaCD91,bonemarrowmDCsandpDCsmature and are able to activate tumor-specific CD8 1 T cells. However, by interacting directly with CD28onlive(nonapoptotic)tumorplasmacells,bonemarrowmDCsdownregulatetheexpressionofproteasomesubunitsinthesecells, thus enabling their evasion from human leukocyte antigen (HLA) class I–restricted CD8 1 T-cell killing. These results suggest that DCs playadual,butopposing,roleinMM:forone,DCsactivateCD8 1 Tcellsagainsttumorplasmacellsand,fortheother,DCsprotecttumor plasmacellsfromCD8 1 T-cellkilling.Thisinformationshouldbetakenintoaccountindesigningimmunotherapyapproachestoenhance immune surveillance in MGUS and to break down immune tolerance in MM. ( Blood . 2015;126(12):1443-1451) different ways of measuring DC activity have produced data 20-26 that cannot be compared and that do not often MGUS patients. Our paper provides a sharper picture of the distribution and func-tioning of DCs during the MGUS-to-MM progression. It shows that a considerable pool of mDCs and pDCs is “ stockpiled ” in BM in MGUS patients and even more so in MM patients. Marrow-resident mDCs and pDCs are fully capable of maturing and activating tumor-speci fi c CD8 1 T cells upon engulfment of apoptotic tumor plasma cells that present calreticulin on their surface. mDCs are also able to interact directly with live tumor plasma cells, downregulate the expression of proteasome subunits in these cells, and make them resistant to CD8 1 T-cell killing.

[1]  P. L. Bergsagel,et al.  CD28-mediated pro-survival signaling induces chemotherapeutic resistance in multiple myeloma. , 2014, Blood.

[2]  F. Dammacco,et al.  MHC class I antigen processing and presenting machinery: organization, function, and defects in tumor cells. , 2013, Journal of the National Cancer Institute.

[3]  O. Joffre,et al.  Cross-presentation by dendritic cells , 2012, Nature Reviews Immunology.

[4]  Je-Jung Lee,et al.  Immunotherapy Using Dendritic Cells against Multiple Myeloma: How to Improve? , 2012, Clinical & developmental immunology.

[5]  P. L. Bergsagel,et al.  CD28 Expressed on Malignant Plasma Cells Induces a Prosurvival and Immunosuppressive Microenvironment , 2011, The Journal of Immunology.

[6]  H. Ueno,et al.  Dendritic cells and immunity against cancer , 2011, Journal of internal medicine.

[7]  Ash A. Alizadeh,et al.  Calreticulin Is the Dominant Pro-Phagocytic Signal on Multiple Human Cancers and Is Counterbalanced by CD47 , 2010, Science Translational Medicine.

[8]  Silvano Sozzani,et al.  Nomenclature of monocytes and dendritic cells in blood. , 2010, Blood.

[9]  F. Dammacco,et al.  Alterations in the antigen processing-presenting machinery of transformed plasma cells are associated with reduced recognition by CD8+ T cells and characterize the progression of MGUS to multiple myeloma. , 2010, Blood.

[10]  Hongbo Zhao,et al.  Phosphatidylinositol 3-kinase/protein kinase B pathway stabilizes DNA methyltransferase I protein and maintains DNA methylation. , 2007, Cellular signalling.

[11]  F. Dammacco,et al.  Bone Marrow of Persistently Hepatitis C Virus-Infected Individuals Accumulates Memory CD8+ T Cells Specific for Current and Historical Viral Antigens: A Study in Patients with Benign Hematological Disorders1 , 2007, The Journal of Immunology.

[12]  S. Jagannath,et al.  Enhancement of clonogenicity of human multiple myeloma by dendritic cells , 2006, The Journal of experimental medicine.

[13]  H. Johnsen,et al.  Impaired functionality and phenotypic profile of dendritic cells from patients with multiple myeloma , 2006, Clinical and experimental immunology.

[14]  W. Janssen,et al.  Cell-Surface Calreticulin Initiates Clearance of Viable or Apoptotic Cells through trans-Activation of LRP on the Phagocyte , 2005, Cell.

[15]  R. Pope,et al.  The evolving clinical profile of abatacept (CTLA4–Ig): a novel co-stimulatory modulator for the treatment of rheumatoid arthritis , 2005, Arthritis research & therapy.

[16]  H. Johnsen,et al.  Impaired circulating myeloid DCs from myeloma patients. , 2004, Cytotherapy.

[17]  M. Dhodapkar,et al.  Vigorous Premalignancy-specific Effector T Cell Response in the Bone Marrow of Patients with Monoclonal Gammopathy , 2003, The Journal of experimental medicine.

[18]  M. Feuerer,et al.  T-cell priming in bone marrow: the potential for long-lasting protective anti-tumor immunity. , 2003, Trends in molecular medicine.

[19]  M. Feuerer,et al.  Bone marrow as a priming site for T-cell responses to blood-borne antigen , 2003, Nature Medicine.

[20]  M. Dhodapkar,et al.  T cells from the tumor microenvironment of patients with progressive myeloma can generate strong, tumor-specific cytolytic responses to autologous, tumor-loaded dendritic cells , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[21]  M. Baccarani,et al.  Dendritic cells are functionally defective in multiple myeloma: the role of interleukin-6. , 2002, Blood.

[22]  D. Joshua,et al.  Dendritic cells from patients with myeloma are numerically normal but functionally defective as they fail to up-regulate CD80 (B7-1) expression after huCD40LT stimulation because of inhibition by transforming growth factor-beta1 and interleukin-10. , 2001, Blood.

[23]  P. Srivastava,et al.  CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. , 2001, Immunity.

[24]  Dongshu Chen,et al.  Bone marrow and peripheral blood dendritic cells from patients with multiple myeloma are phenotypically and functionally normal despite the detection of Kaposi's sarcoma herpesvirus gene sequences. , 1999, Blood.

[25]  H. Goldschmidt,et al.  Dendritic cells generated from the blood of patients with multiple myeloma are phenotypically and functionally identical to those similarly produced from healthy donors , 1997, British journal of haematology.

[26]  P. Doherty,et al.  Bone marrow can function as a lymphoid organ during a primary immune response under conditions of disrupted lymphocyte trafficking. , 1997, Journal of immunology.

[27]  Patrizia Agostinis,et al.  Immunogenic cell death, DAMPs and anticancer therapeutics: an emerging amalgamation. , 2010, Biochimica et biophysica acta.

[28]  M. Pérez‐Andrés,et al.  Peripheral blood dendritic cell subsets from patients with monoclonal gammopathies show an abnormal distribution and are functionally impaired. , 2008, The oncologist.

[29]  A. Palucka,et al.  Interactions of tumor cells with dendritic cells: balancing immunity and tolerance , 2008, Cell Death and Differentiation.

[30]  L. Zitvogel,et al.  Calreticulin exposure dictates the immunogenicity of cancer cell death , 2007, Nature Medicine.

[31]  Laurence Zitvogel,et al.  Antigen presentation and T cell stimulation by dendritic cells. , 2002, Annual review of immunology.

[32]  O. Cope,et al.  Multiple myeloma. , 1948, The New England journal of medicine.