Abstract
Castleman disease (CD), a heterogeneous group of disorders that share morphological features, is divided into unicentric CD and multicentric CD (MCD) according to the clinical presentation and disease course. Unicentric CD involves a solitary enlarged lymph node and mild symptoms and excision surgery is often curative. MCD includes a form associated with Kaposi sarcoma herpesvirus (KSHV) (also known as human herpesvirus 8) and a KSHV-negative idiopathic form (iMCD). iMCD can present in association with severe syndromes such as TAFRO (thrombocytopenia, ascites, fever, reticulin fibrosis and organomegaly) or POEMS (polyneuropathy, organomegaly, endocrinopathy, monoclonal plasma cell disorder and skin changes). KSHV-MCD often occurs in the setting of HIV infection or another cause of immune deficiency. The interplay between KSHV and HIV elevates the risk for the development of KSHV-induced disorders, including KSHV-MCD, KSHV-lymphoproliferation, KSHV inflammatory cytokine syndrome, primary effusion lymphoma and Kaposi sarcoma. A CD diagnosis requires a multidimensional approach, including clinical presentation and imaging, pathological features, and molecular virology. B cell-directed monoclonal antibody therapy is the standard of care in KSHV-MCD, and anti-IL-6 therapy is the recommended first-line therapy and only treatment of iMCD approved by the US FDA and EMA.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 1 digital issues and online access to articles
$119.00 per year
only $119.00 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
References
IARC. A Review of Human Carcinogens. Part B: Biological Agents Vol. 100 (IARC, 2012).
Castleman, B. & Towne, V. W. Case records of the Massachusetts General Hospital: Case No. 40231. N. Engl. J. Med. 250, 1001–1005 (1954).
Keller, A. R., Hochholzer, L. & Castleman, B. Hyaline-vascular and plasma-cell types of giant lymph node hyperplasia of the mediastinum and other locations. Cancer 29, 670–683 (1972).
Kessler, E. Multicentric giant lymph node hyperplasia. A report of seven cases. Cancer 56, 2446–2451 (1985).
van Rhee, F. et al. International evidence-based consensus diagnostic and treatment guidelines for unicentric Castleman disease. Blood Adv. 4, 6039–6050 (2020).
Soulier, J. et al. Kaposi’s sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman’s disease. Blood 86, 1276–1280 (1995).
Carbone, A., De Paoli, P., Gloghini, A. & Vaccher, E. KSHV-associated multicentric Castleman disease: A tangle of different entities requiring multitarget treatment strategies. Int. J. Cancer 137, 251–261 (2015). This review focuses on the aetiopathogenic role of co-infection by herpesviruses (KSHV and EBV) in PLWH.
Ramaswami, R. et al. Characteristics and outcomes of KSHV-associated multicentric Castleman disease with or without other KSHV diseases. Blood Adv. 5, 1660–1670 (2021).
Carbone, A., Cesarman, E., Spina, M., Gloghini, A. & Schulz, T. F. HIV-associated lymphomas and gamma-herpesviruses. Blood 113, 1213–1224 (2009).
Polizzotto, M. N., Uldrick, T. S., Hu, D. & Yarchoan, R. Clinical manifestations of Kaposi sarcoma herpesvirus lytic activation: multicentric Castleman disease (KSHV-MCD) and the KSHV inflammatory cytokine syndrome. Front. Microbiol. 3, 73 (2012).
Kojima, M. et al. Clinical implications of idiopathic multicentric Castleman disease among Japanese: a report of 28 cases. Int. J. Surg. Pathol. 16, 391–398 (2008). The paper suggests that at variance with iMCD in the Western world, the chronic course of the disease in Japan appears to be related to negativity for HHV8 infection.
Bélec, L. et al. Human herpesvirus 8 infection in patients with POEMS syndrome-associated multicentric Castleman’s disease. Blood 93, 3643–3653 (1999).
Vega, F., Miranda, R. N. & Medeiros, L. J. KSHV/HHV8-positive large B-cell lymphomas and associated diseases: a heterogeneous group of lymphoproliferative processes with significant clinicopathological overlap. Mod. Pathol. 33, 18–28 (2020). This review focuses on the current diagnosis of HHV8-associated lymphoproliferative disorders ranging from MCD to frank lymphomas.
Bower, M. How I treat HIV-associated multicentric Castleman disease. Blood 116, 4415–4421 (2010).
Dispenzieri, A. & Fajgenbaum, D. C. Overview of Castleman disease. Blood 135, 1353–1364 (2020). The advent of effective retroviral therapy and the use of rituximab in HHV8-MCD have improved outcomes in HHV8-MCD. Anti-IL-6-directed therapies are highly effective in many iMCD patients but additional therapies are required for refractory cases.
Gopal, S. et al. Multicentric Castleman’s disease in Malawi. Lancet 384, 1158 (2014).
Cohen, A. B. et al. Clinical characteristics, treatment patterns, and overall survival of real-world patients with idiopathic multicentric Castleman disease. J. Clin. Oncol. 39, 7048–7048 (2021).
Simpson, D. Epidemiology of Castleman disease. Hematol. Oncol. Clin. North. Am. 32, 1–10 (2018).
Oksenhendler, E. et al. The full spectrum of Castleman disease: 273 patients studied over 20 years. Br. J. Haematol. 180, 206–216 (2018). The study demonstrates that, although patients with iMCD and HHV8-associated MCD displayed similar clinical characteristics, the disease was more aggressive in HHV8-associated MCD, particularly in PLWH.
van Rhee, F. et al. International, evidence-based consensus treatment guidelines for idiopathic multicentric Castleman disease. Blood 132, 2115–2124 (2018). The purpose of this paper is to establish a consensus for treatment guidelines based on the severity of iMCD.
Murakami, M. et al. Clinicopathologic characteristics of 342 patients with multicentric Castleman disease in Japan. Mod. Rheumatol. 30, 843–851 (2020).
Gopal, S. et al. Characteristics and survival for HIV-associated multicentric Castleman disease in Malawi. J. Int. AIDS Soc. 18, 20122 (2015).
Patel, M. et al. in Immunopathology and Immunomodulation (ed. Metodiev, K.) 247–259 (IntechOpen, 2015).
Parez, N., Bader-Meunier, B., Roy, C. C. & Dommergues, J. P. Paediatric Castleman disease: report of seven cases and review of the literature. Eur. J. Pediatr. 158, 631–637 (1999).
Borocco, C. et al. The French paediatric cohort of Castleman disease: a retrospective report of 23 patients. Orphanet J. Rare Dis. 15, 95 (2020). Unlike adult CD, which may be associated with HIV and HHV8 infection, paediatric CD could be favoured by primary activation of innate immunity and may affect life expectancy less.
Leroy, S. et al. Multicentric Castleman disease in an HHV8-infected child born to consanguineous parents with systematic review. Pediatrics 129, e199–e203 (2012).
Masaki, Y. et al. Epidemiological analysis of multicentric and unicentric Castleman disease and TAFRO syndrome in Japan. J. Clin. Exp. Hematop. 59, 175–178 (2019).
Munshi, N. et al. Use of a claims database to characterize and estimate the incidence rate for Castleman disease. Leuk. Lymphoma 56, 1252–1260 (2015).
Iftode, N., Rădulescu, M. A., Aramă, Ș. S. & Aramă, V. Update on Kaposi sarcoma-associated herpesvirus (KSHV or HHV8) — review. Rom. J. Intern. Med. 58, 199–208 (2020).
Chihara, D. et al. Differences in incidence and trends of haematological malignancies in Japan and the United States. Br. J. Haematol. 164, 536–545 (2014).
Powles, T. et al. The role of immune suppression and HHV-8 in the increasing incidence of HIV-associated multicentric Castleman’s disease. Ann. Oncol. 20, 775–779 (2009).
Zhang, L. et al. Clinical spectrum and survival analysis of 145 cases of HIV-negative Castleman’s disease: renal function is an important prognostic factor. Sci. Rep. 6, 23831 (2016).
Gonçalves, P. H., Uldrick, T. S. & Yarchoan, R. HIV-associated Kaposi sarcoma and related diseases. AIDS 31, 1903–1916 (2017).
Cesarman, E. et al. Kaposi sarcoma. Nat. Rev. Dis. Prim. 5, 9 (2019).
Mesri, E. A., Cesarman, E. & Boshoff, C. Kaposi’s sarcoma and its associated herpesvirus. Nat. Rev. Cancer 10, 707–719 (2010).
Dispenzieri, A. et al. The clinical spectrum of Castleman’s disease. Am. J. Hematol. 87, 997–1002 (2012). The paper supports the concept of four categories of CD, namely (1) unicentric CD (91%), (2) multicentric CD associated with the osteosclerotic variant of POEMS syndrome (90%), (3) multicentric CD without POEMS syndrome (65%), and (4) multicentric CD with POEMS syndrome without osteosclerotic lesions (27%).
Weisenburger, D. D., Nathwani, B. N., Winberg, C. D. & Rappaport, H. Multicentric angiofollicular lymph node hyperplasia: a clinicopathologic study of 16 cases. Hum. Pathol. 16, 162–172 (1985).
Chang, K. C. et al. Monoclonality and cytogenetic abnormalities in hyaline vascular Castleman disease. Mod. Pathol. 27, 823–831 (2014).
Li, Z. et al. Recurrent PDGFRB mutations in unicentric Castleman disease. Leukemia 33, 1035–1038 (2019).
Nabel, C. S. et al. Virome capture sequencing does not identify active viral infection in unicentric and idiopathic multicentric Castleman disease. PLoS ONE 14, e0218660 (2019).
Fajgenbaum, D. C., van Rhee, F. & Nabel, C. S. HHV-8-negative, idiopathic multicentric Castleman disease: novel insights into biology, pathogenesis, and therapy. Blood 123, 2924–2933 (2014).
Goodman, A. M. et al. Novel somatic alterations in unicentric and idiopathic multicentric castleman disease. Eur. J. Haematol. https://doi.org/10.1111/ejh.13702 (2021).
Nagy, A. et al. Next-generation sequencing of idiopathic multicentric and unicentric Castleman disease and follicular dendritic cell sarcomas. Blood Adv. 2, 481–491 (2018).
Lim, J. Y. et al. DNMT3A haploinsufficiency causes dichotomous DNA methylation defects at enhancers in mature human immune cells. J. Exp. Med. 218, e20202733 (2021).
Yoshimi, A. et al. Genetic basis for iMCD-TAFRO. Oncogene 39, 3218–3225 (2020).
Baker, T. S. et al. A novel FAS mutation with variable expressivity in a family with unicentric and idiopathic multicentric Castleman disease. Blood Adv. 2, 2959–2963 (2018).
Fajgenbaum, D. C. & June, C. H. Cytokine storm. N. Engl. J. Med. 383, 2255–2273 (2020). The review describes the role of the cytokine storm in MCD.
van Rhee, F., Stone, K., Szmania, S., Barlogie, B. & Singh, Z. Castleman disease in the 21st century: an update on diagnosis, assessment, and therapy. Clin. Adv. Hematol. Oncol. 8, 486–498 (2010).
van Gameren, M. M. et al. Effects of recombinant human interleukin-6 in cancer patients: a phase I-II study. Blood 84, 1434–1441 (1994).
Gherardi, R. K. et al. Elevated levels of interleukin-1 beta (IL-1 beta) and IL-6 in serum and increased production of IL-1 beta mRNA in lymph nodes of patients with polyneuropathy, organomegaly, endocrinopathy, M protein, and skin changes (POEMS) syndrome. Blood 83, 2587–2593 (1994).
Lee, M. et al. Multicentric Castleman’s disease with an increased serum level of macrophage colony-stimulating factor. Am. J. Hematol. 54, 321–323 (1997).
Nishi, J. et al. Expression of vascular endothelial growth factor in sera and lymph nodes of the plasma cell type of Castleman’s disease. Br. J. Haematol. 104, 482–485 (1999).
Pierson, S. K. et al. ACCELERATE: a patient-powered natural history study design enabling clinical and therapeutic discoveries in a rare disorder. Cell Rep. Med. 1, 100158 (2020).
Pierson, S. K. et al. Plasma proteomics identifies a ‘chemokine storm’ in idiopathic multicentric Castleman disease. Am. J. Hematol. 93, 902–912 (2018).
Fajgenbaum, D. C. et al. Identifying and targeting pathogenic PI3K/AKT/mTOR signaling in IL-6-blockade-refractory idiopathic multicentric Castleman disease. J. Clin. Invest. 129, 4451–4463 (2019).
An, J., Lichtenstein, A. K., Brent, G. & Rettig, M. B. The Kaposi sarcoma-associated herpesvirus (KSHV) induces cellular interleukin 6 expression: role of the KSHV latency-associated nuclear antigen and the AP1 response element. Blood 99, 649–654 (2002).
Pai, R. L. et al. Type I IFN response associated with mTOR activation in the TAFRO subtype of idiopathic multicentric Castleman disease. JCI Insight 5, e135031 (2020).
Pierson, S. K. et al. Discovery and validation of a novel subgroup and therapeutic target in idiopathic multicentric Castleman disease. Blood Adv. 5, 3445–3456 (2021).
Arenas, D. J. et al. Increased mTOR activation in idiopathic multicentric Castleman disease. Blood 135, 1673–1684 (2020).
Endo, Y. et al. Mediterranean fever gene variants modify clinical phenotypes of idiopathic multi-centric Castleman disease. Clin. Exp. Immunol. 206, 91–98 (2021).
Endo, Y. et al. Idiopathic multicentric Castleman disease with novel heterozygous Ile729Met mutation in exon 10 of familial Mediterranean fever gene. Rheumatology 60, 445–450 (2021).
Endo, Y. et al. Successful canakinumab treatment for activated innate response in idiopathic Castleman’s disease with multiple heterozygous MEFV exon 2 variants. Clin. Immunol. 219, 108547 (2020).
Du, M. Q. et al. Kaposi sarcoma-associated herpesvirus infects monotypic (IgM lambda) but polyclonal naive B cells in Castleman disease and associated lymphoproliferative disorders. Blood 97, 2130–2136 (2001).
Dupin, N. et al. Distribution of human herpesvirus-8 latently infected cells in Kaposi’s sarcoma, multicentric Castleman’s disease, and primary effusion lymphoma. Proc. Natl Acad. Sci. USA 96, 4546–4551 (1999).
Parravicini, C. et al. Differential viral protein expression in Kaposi’s sarcoma-associated herpesvirus-infected diseases: Kaposi’s sarcoma, primary effusion lymphoma, and multicentric Castleman’s disease. Am. J. Pathol. 156, 743–749 (2000).
Totonchy, J. et al. KSHV induces immunoglobulin rearrangements in mature B lymphocytes. PLoS Pathog. 14, e1006967 (2018).
Jacobs, S. R. et al. The viral interferon regulatory factors of kaposi’s sarcoma-associated herpesvirus differ in their inhibition of interferon activation mediated by toll-like receptor 3. J. Virol. 87, 798–806 (2013).
Dittmer, D. P. & Damania, B. Kaposi sarcoma-associated herpesvirus: immunobiology, oncogenesis, and therapy. J. Clin. Invest. 126, 3165–3175 (2016).
Uldrick, T. S. et al. High-dose zidovudine plus valganciclovir for Kaposi sarcoma herpesvirus-associated multicentric Castleman disease: a pilot study of virus-activated cytotoxic therapy. Blood 117, 6977–6986 (2011).
Bhatt, A. P. et al. A viral kinase mimics S6 kinase to enhance cell proliferation. Proc. Natl Acad. Sci. USA 113, 7876–7881 (2016).
Anders, P. M. et al. Human herpesvirus-encoded kinase induces B cell lymphomas in vivo. J. Clin. Invest. 128, 2519–2534 (2018).
Cannon, J. S. et al. Heterogeneity of viral IL-6 expression in HHV-8-associated diseases. J. Infect. Dis. 180, 824–828 (1999).
Parravicini, C. et al. Expression of a virus-derived cytokine, KSHV vIL-6, in HIV-seronegative Castleman’s disease. Am. J. Pathol. 151, 1517–1522 (1997).
Staskus, K. A. et al. Cellular tropism and viral interleukin-6 expression distinguish human herpesvirus 8 involvement in Kaposi’s sarcoma, primary effusion lymphoma, and multicentric Castleman’s disease. J. Virol. 73, 4181–4187 (1999).
Polizzotto, M. N. et al. Human and viral interleukin-6 and other cytokines in Kaposi sarcoma herpesvirus-associated multicentric Castleman disease. Blood 122, 4189–4198 (2013).
Oksenhendler, E. et al. High levels of human herpesvirus 8 viral load, human interleukin-6, interleukin-10, and C reactive protein correlate with exacerbation of multicentric Castleman disease in HIV-infected patients. Blood 96, 2069–2073 (2000).
Aoki, Y., Tosato, G., Fonville, T. W. & Pittaluga, S. Serum viral interleukin-6 in AIDS-related multicentric Castleman disease. Blood 97, 2526–2527 (2001).
An, J., Sun, Y., Sun, R. & Rettig, M. B. Kaposi’s sarcoma-associated herpesvirus encoded vFLIP induces cellular IL-6 expression: the role of the NF-kappaB and JNK/AP1 pathways. Oncogene 22, 3371–3385 (2003).
Deng, H., Chu, J. T., Rettig, M. B., Martinez-Maza, O. & Sun, R. Rta of the human herpesvirus 8/Kaposi sarcoma-associated herpesvirus up-regulates human interleukin-6 gene expression. Blood 100, 1919–1921 (2002).
Chadburn, A. et al. HHV8/KSHV-positive lymphoproliferative disorders and the spectrum of plasmablastic and plasma cell neoplasms: 2015 SH/EAHP Workshop Report-Part 3. Am. J. Clin. Pathol. 147, 171–187 (2017).
Gasperini, P., Sakakibara, S. & Tosato, G. Contribution of viral and cellular cytokines to Kaposi’s sarcoma-associated herpesvirus pathogenesis. J. Leukoc. Biol. 84, 994–1000 (2008).
Suda, T. et al. HHV-8 infection status of AIDS-unrelated and AIDS-associated multicentric Castleman’s disease. Pathol. Int. 51, 671–679 (2001).
Kawabata, H. et al. Clinical features and treatment of multicentric Castleman’s disease: a retrospective study of 21 Japanese patients at a single institute. J. Clin. Exp. Hematop. 53, 69–77 (2013).
Tosato, G. et al. Monocyte-derived human B-cell growth factor identified as interferon-beta 2 (BSF-2, IL-6). Science 239, 502–504 (1988).
Murakami, M. et al. Critical cytoplasmic region of the interleukin 6 signal transducer gp130 is conserved in the cytokine receptor family. Proc. Natl Acad. Sci. USA 88, 11349–11353 (1991).
Nicholas, J. et al. Kaposi’s sarcoma-associated human herpesvirus-8 encodes homologues of macrophage inflammatory protein-1 and interleukin-6. Nat. Med. 3, 287–292 (1997).
Chatterjee, M., Osborne, J., Bestetti, G., Chang, Y. & Moore, P. S. Viral IL-6-induced cell proliferation and immune evasion of interferon activity. Science 298, 1432–1435 (2002).
Giffin, L. & Damania, B. KSHV: pathways to tumorigenesis and persistent infection. Adv. Virus Res. 88, 111–159 (2014).
Giffin, L., West, J.A. & Damania, B. Kaposi’s sarcoma-associated herpesvirus interleukin-6 modulates endothelial cell movement by upregulating cellular genes involved in migration. mBio 6, e01499-15 (2015).
Nishi, J. & Maruyama, I. Increased expression of vascular endothelial growth factor (VEGF) in Castleman’s disease: proposed pathomechanism of vascular proliferation in the affected lymph node. Leuk. Lymphoma 38, 387–394 (2000).
Aoki, Y. et al. Angiogenesis and hematopoiesis induced by Kaposi’s sarcoma-associated herpesvirus-encoded interleukin-6. Blood 93, 4034–4043 (1999).
Brousset, P., Cesarman, E., Meggetto, F., Lamant, L. & Delsol, G. Colocalization of the viral interleukin-6 with latent nuclear antigen-1 of human herpesvirus-8 in endothelial spindle cells of Kaposi’s sarcoma and lymphoid cells of multicentric Castleman’s disease. Hum. Pathol. 32, 95–100 (2001).
McNamara, R. P. et al. Extracellular vesicles from Kaposi sarcoma-associated herpesvirus lymphoma induce long-term endothelial cell reprogramming. PLoS Pathog. 15, e1007536 (2019).
O’Hara, A. J. et al. Pre-micro RNA signatures delineate stages of endothelial cell transformation in Kaposi sarcoma. PLoS Pathog. 5, e1000389 (2009).
Said, J. et al. in WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues (eds Swerdlow, S. H. et al) 325–329 (IARC, 2017).
Moore, P. S., Boshoff, C., Weiss, R. A. & Chang, Y. Molecular mimicry of human cytokine and cytokine response pathway genes by KSHV. Science 274, 1739–1744 (1996).
Sarek, G. et al. Reactivation of the p53 pathway as a treatment modality for KSHV-induced lymphomas. J. Clin. Invest. 117, 1019–1028 (2007).
Uldrick, T. S. et al. An interleukin-6-related systemic inflammatory syndrome in patients co-infected with Kaposi sarcoma-associated herpesvirus and HIV but without multicentric Castleman disease. Clin. Infect. Dis. 51, 350–358 (2010).
Caro-Vegas, C. et al. Runaway Kaposi sarcoma-associated herpesvirus replication correlates with systemic IL-10 levels. Virology 539, 18–25 (2020).
Tamburro, K. M. et al. Vironome of Kaposi sarcoma associated herpesvirus-inflammatory cytokine syndrome in an AIDS patient reveals co-infection of human herpesvirus 8 and human herpesvirus 6A. Virology 433, 220–225 (2012).
Boutboul, D. et al. Treatment and outcome of unicentric Castleman disease: a retrospective analysis of 71 cases. Br. J. Haematol. 186, 269–273 (2019).
Zhang, M. Y. et al. UCD with MCD-like inflammatory state: surgical excision is highly effective. Blood Adv. 5, 122–128 (2021).
Zhen, J. F. et al. Clinical analysis of unicentric Castleman’s disease with paraneoplastic pemphigus and bronchiolitis obliterans. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 39, 492–498 (2017).
Fajgenbaum, D. C. et al. International, evidence-based consensus diagnostic criteria for HHV-8-negative/idiopathic multicentric Castleman disease. Blood 129, 1646–1657 (2017).
Teruya-Feldstein, J. et al. Expression of human herpesvirus-8 oncogene and cytokine homologues in an HIV-seronegative patient with multicentric Castleman’s disease and primary effusion lymphoma. Lab. Invest. 78, 1637–1642 (1998).
Haq, I. U. et al. The clinical application of plasma Kaposi sarcoma herpesvirus viral load as a tumour biomarker: results from 704 patients. HIV Med. 17, 56–61 (2016).
Polizzotto, M. N. et al. Clinical features and outcomes of patients with symptomatic Kaposi sarcoma herpesvirus (KSHV)-associated inflammation: prospective characterization of KSHV inflammatory cytokine syndrome (KICS). Clin. Infect. Dis. 62, 730–738 (2016). KICS may be an unrecognized cause of morbidity and mortality in patients with KSHV-MCD.
Lurain, K. et al. Viral, immunologic, and clinical features of primary effusion lymphoma. Blood 133, 1753–1761 (2019).
Fujimoto, S. et al. Tentative diagnostic criteria and disease severity classification for Castleman disease: a report of the research group on Castleman disease in Japan. Mod. Rheumatol. 28, 161–167 (2018).
Iwaki, N. et al. Elevated serum interferon gamma-induced protein 10 kDa is associated with TAFRO syndrome. Sci. Rep. 7, 42316 (2017).
Polizzotto, M. N. et al. 18F-fluorodeoxyglucose positron emission tomography in Kaposi sarcoma herpesvirus-associated multicentric Castleman disease: correlation with activity, severity, inflammatory and virologic parameters. J. Infect. Dis. 212, 1250–1260 (2015).
Gloghini, A., Dolcetti, R. & Carbone, A. Lymphomas occurring specifically in HIV-infected patients: from pathogenesis to pathology. Semin. Cancer Biol. 23, 457–467 (2013).
Chadburn, A. et al. Immunophenotypic analysis of the Kaposi sarcoma herpesvirus (KSHV; HHV-8)-infected B cells in HIV+ multicentric Castleman disease (MCD). Histopathology 53, 513–524 (2008).
Oksenhendler, E. et al. Multicentric Castleman’s disease in HIV infection: a clinical and pathological study of 20 patients. AIDS 10, 61–67 (1996).
Cronin, D. M. & Warnke, R. A. Castleman disease: an update on classification and the spectrum of associated lesions. Adv. Anat. Pathol. 16, 236–246 (2009).
Naresh, K. N., Trivedi, P., Horncastle, D. & Bower, M. CD20 expression in the HHV-8-infected lymphoid cells in multicentric Castleman disease. Histopathology 55, 358–359 (2009).
Naresh, K. N., Rice, A. J. & Bower, M. Lymph nodes involved by multicentric Castleman disease among HIV-positive individuals are often involved by Kaposi sarcoma. Am. J. Surg. Pathol. 32, 1006–1012 (2008).
Liu, A. Y. et al. Idiopathic multicentric Castleman’s disease: a systematic literature review. Lancet Haematol. 3, e163–e175 (2016).
Stone, K. et al. Interleukin-6 receptor polymorphism is prevalent in HIV-negative Castleman disease and is associated with increased soluble interleukin-6 receptor levels. PLoS ONE 8, e54610 (2013).
Fayand, A. et al. Epidemiology of Castleman disease associated with AA amyloidosis: description of 2 new cases and literature review. Amyloid 26, 197–202 (2019).
Ohzono, A. et al. Clinical and immunological findings in 104 cases of paraneoplastic pemphigus. Br. J. Dermatol. 173, 1447–1452 (2015).
Raza, H. A. et al. Unicentric Castleman disease complicated by paraneoplastic bronchiolitis obliterans and pemphigus. Respir. Med. Case Rep. 25, 129–132 (2018).
Gerald, W., Kostianovsky, M. & Rosai, J. Development of vascular neoplasia in Castleman’s disease. Report of seven cases. Am. J. Surg. Pathol. 14, 603–614 (1990).
Castleman, B., Iverson, L. & Menendez, V. Localized mediastinal lymph-node hyperplasia resembling thymoma. Cancer 9, 822–830 (1956).
Lachmann, H. J., Gilbertson, J. A., Gillmore, J. D., Hawkins, P. N. & Pepys, M. B. Unicentric Castleman’s disease complicated by systemic AA amyloidosis: a curable disease. QJM 95, 211–218 (2002).
Yoshizaki, K. et al. Pathogenic significance of interleukin-6 (IL-6/BSF-2) in Castleman’s disease. Blood 74, 1360–1367 (1989).
van Rhee, F. et al. Siltuximab for multicentric Castleman’s disease: a randomised, double-blind, placebo-controlled trial. Lancet Oncol. 15, 966–974 (2014).
Casper, C. et al. Analysis of inflammatory and anemia-related biomarkers in a randomized, double-blind, placebo-controlled study of siltuximab (anti-IL6 monoclonal antibody) in patients with multicentric Castleman disease. Clin. Cancer Res. 21, 4294–4304 (2015).
Nishimoto, N. et al. Humanized anti-interleukin-6 receptor antibody treatment of multicentric Castleman disease. Blood 106, 2627–2632 (2005).
Morra, D. E. et al. Predictors of response to anti-IL6 monoclonal antibody therapy (siltuximab) in idiopathic multicentric Castleman disease: secondary analyses of phase II clinical trial data. Br. J. Haematol. 184, 232–241 (2019).
Fajgenbaum, D. C. et al. Insufficient evidence exists to use histopathologic subtype to guide treatment of idiopathic multicentric Castleman disease. Am. J. Hematol. 95, 1553–1561 (2020).
Gholam, D., Vantelon, J. M., Al-Jijakli, A. & Bourhis, J. H. A case of multicentric Castleman’s disease associated with advanced systemic amyloidosis treated with chemotherapy and anti-CD20 monoclonal antibody. Ann. Hematol. 82, 766–768 (2003).
Ocio, E. M. et al. Efficacy of rituximab in an aggressive form of multicentric Castleman disease associated with immune phenomena. Am. J. Hematol. 78, 302–305 (2005).
Ide, M., Kawachi, Y., Izumi, Y., Kasagi, K. & Ogino, T. Long-term remission in HIV-negative patients with multicentric Castleman’s disease using rituximab. Eur. J. Haematol. 76, 119–123 (2006).
Mian, H. & Leber, B. Mixed variant multicentric Castleman disease treated with rituximab: case report. J. Pediatr. Hematol. Oncol. 32, 622 (2010).
Chen, L. Y. C., Skinnider, B. F., Wilson, D. & Fajgenbaum, D. C. Adrenalitis and anasarca in idiopathic multicentric Castleman’s disease. Lancet 397, 1749 (2021).
Akiyama, M., Kaneko, Y. & Takeuchi, T. Tocilizumab for the treatment of TAFRO syndrome: a systematic literature review. Ann. Hematol. 99, 2463–2475 (2020).
Fujimoto, S. et al. Optimal treatments for TAFRO syndrome: a retrospective surveillance study in Japan. Int. J. Hematol. 113, 73–80 (2021).
Masaki, Y. et al. 2019 Updated diagnostic criteria and disease severity classification for TAFRO syndrome. Int. J. Hematol. 111, 155–158 (2020).
Dispenzieri, A. How I treat POEMS syndrome. Blood 119, 5650–5658 (2012).
Kuwabara, S., Dispenzieri, A., Arimura, K., Misawa, S. & Nakaseko, C. Treatment for POEMS (polyneuropathy, organomegaly, endocrinopathy, M-protein, and skin changes) syndrome. Cochrane Database Syst. Rev. 2012, CD006828 (2012).
Gavriatopoulou, M. et al. European Myeloma Network recommendations on diagnosis and management of patients with rare plasma cell dyscrasias. Leukemia 32, 1883–1898 (2018).
Gerard, L. et al. Prospective study of rituximab in chemotherapy-dependent human immunodeficiency virus associated multicentric Castleman’s disease: ANRS 117 CastlemaB Trial. J. Clin. Oncol. 25, 3350–3356 (2007).
Bower, M. et al. Brief communication: rituximab in HIV-associated multicentric Castleman disease. Ann. Intern. Med. 147, 836–839 (2007).
Hoffmann, C. et al. Improved outcome with rituximab in patients with HIV-associated multicentric Castleman disease. Blood 118, 3499–3503 (2011).
Gerard, L. et al. Rituximab decreases the risk of lymphoma in patients with HIV-associated multicentric Castleman disease. Blood 119, 2228–2233 (2012).
Pria, A. D. et al. Relapse of HHV8-positive multicentric Castleman disease following rituximab-based therapy in HIV-positive patients. Blood 129, 2143–2147 (2017).
Casper, C., Nichols, W. G., Huang, M. L., Corey, L. & Wald, A. Remission of HHV-8 and HIV-associated multicentric Castleman disease with ganciclovir treatment. Blood 103, 1632–1634 (2004).
Berezne, A., Agbalika, F. & Oksenhendler, E. Failure of cidofovir in HIV-associated multicentric Castleman disease. Blood 103, 4368–4369 (2004).
Senanayake, S. et al. Multicentric Castleman’s disease treated with antivirals and immunosuppressants. J. Med. Virol. 71, 399–403 (2003).
Ramaswami, R. et al. Tocilizumab in patients with symptomatic Kaposi sarcoma herpesvirus-associated multicentric Castleman disease. Blood 135, 2316–2319 (2020).
Oksenhendler, E. et al. High incidence of Kaposi sarcoma-associated herpesvirus-related non-Hodgkin lymphoma in patients with HIV infection and multicentric Castleman disease. Blood 99, 2331–2336 (2002).
Marcelin, A. G. et al. Rituximab therapy for HIV-associated Castleman disease long-term remission of Kaposi sarcoma-associated herpesvirus-related multicentric Castleman disease with anti-CD20 monoclonal antibody therapy. Blood 102, 2786–2788 (2003).
Karass, M., Grossniklaus, E., Seoud, T., Jain, S. & Goldstein, D. A. Kaposi sarcoma inflammatory cytokine syndrome (KICS): a rare but potentially treatable condition. Oncologist 22, 623–625 (2017).
Talat, N., Belgaumkar, A. P. & Schulte, K. M. Surgery in Castleman’s disease: a systematic review of 404 published cases. Ann. Surg. 255, 677–684 (2012).
Sitenga, J., Aird, G., Ahmed, A. & Silberstein, P. T. Impact of siltuximab on patient-related outcomes in multicentric Castleman’s disease. Patient Relat. Outcome Meas. 9, 35–41 (2018).
van Rhee, F. et al. Patient-reported outcomes for multicentric Castleman’s disease in a randomized, placebo-controlled study of siltuximab. Patient 8, 207–216 (2015).
Vernon, M. et al. Deriving health utility values from a randomized, double-blind, placebo-controlled trial of siltuximab in subjects with multicentric Castleman’s disease. Curr. Med. Res. Opin. 32, 1193–1200 (2016).
Slade, A. et al. Patient reported outcome measures in rare diseases: a narrative review. Orphanet J. Rare Dis. 13, 61 (2018).
Casper, C. et al. The Multicentric Castleman’s Disease (MCD)-Symptom Scale (MCD-SS): development and validation of a patient-reported outcome (PRO) measure for an ultra-orphan disease. Value Health 17, A535 (2014).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02817997 (2016).
Guerit, E., Arts, F., Dachy, G., Boulouadnine, B. & Demoulin, J. B. PDGF receptor mutations in human diseases. Cell Mol. Life Sci. 78, 3867–3881 (2021).
Brandt, S. J., Bodine, D. M., Dunbar, C. E. & Nienhuis, A. W. Dysregulated interleukin 6 expression produces a syndrome resembling Castleman’s disease in mice. J. Clin. Invest. 86, 592–599 (1990).
Suthaus, J. et al. HHV-8-encoded viral IL-6 collaborates with mouse IL-6 in the development of multicentric Castleman disease in mice. Blood 119, 5173–5181 (2012).
Nishimura, Y. et al. Validated international definition of the thrombocytopenia, anasarca, fever, reticulin fibrosis, renal insufficiency, and organomegaly clinical subtype (TAFRO) of idiopathic multicentric Castleman disease. Am. J. Hematol. 96, 1241–1252 (2021).
Wang, H. W., Pittaluga, S. & Jaffe, E. S. Multicentric Castleman disease: where are we now? Semin. Diagn. Pathol. 33, 294–306 (2016).
Gonzalez-Farre, B. et al. HHV8-related lymphoid proliferations: a broad spectrum of lesions from reactive lymphoid hyperplasia to overt lymphoma. Mod. Pathol. 30, 745–760 (2017).
Bower, M. et al. British HIV association guidelines for HIV-associated malignancies 2014. HIV Med. 15, 1–92 (2014).
Olszewski, A. J., Fallah, J. & Castillo, J. J. Human immunodeficiency virus-associated lymphomas in the antiretroviral therapy era: analysis of the National Cancer Data Base. Cancer 122, 2689–2697 (2016).
El-Fattah, M. A. Clinical characteristics and survival outcome of primary effusion lymphoma: a review of 105 patients. Hematol. Oncol. 35, 878–883 (2017).
Shimada, K., Hayakawa, F. & Kiyoi, H. Biology and management of primary effusion lymphoma. Blood 132, 1879–1888 (2018).
Castillo, J. J. et al. Prognostic factors in chemotherapy-treated patients with HIV-associated plasmablastic lymphoma. Oncologist 15, 293–299 (2010).
Acknowledgements
M. Bor. is supported by NIH grants 2UM1CA121947-09, 5U01-I068636 and R25TW011215. B.D. is supported by NIH grants DE028211, CA096500, CA254564, CA239583 and CA163217. M.N.P. is supported by the Australian National Health and Medical Research Council and by the Cancer Institute of New South Wales. The authors would like to thank the patients and their families for sharing their experiences as outlined in Box 6.
Author information
Authors and Affiliations
Contributions
Introduction (A.C.); Epidemiology (M. Bor.); Mechanisms/pathophysiology (B.D. and A.G.); Diagnosis, screening and prevention (M.N.P.); Management (D.C.F. and M. Bow.); Quality of life (R.K.J. and D.C.F.); Outlook (M. Bow.); Overview of Primer (A.C.)
Corresponding author
Ethics declarations
Competing interests
M. Bow. received speaker honoraria from Gilead sciences, BMS, MSD, Janssen and ViiV. M.N.P. received speaker honoraria from Gilead Sciences and research funding from BMS, Celgene, Janssen and ViiV (to the institution, outside the area of the submitted work). D.C.F. has received research funding from EUSA Pharma and Janssen Pharmaceuticals, the study drug from Pfizer for a clinical trial of sirolimus in Castleman disease, and two provisional patents pending on the diagnosis and treatment of Castleman disease. The other authors declare no competing interests.
Additional information
Peer review information
Nature Reviews Disease Primers thanks M. Ide, who co-reviewed with T. Koga; J. Li, T. Schulz; and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
About this article
Cite this article
Carbone, A., Borok, M., Damania, B. et al. Castleman disease. Nat Rev Dis Primers 7, 84 (2021). https://doi.org/10.1038/s41572-021-00317-7
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1038/s41572-021-00317-7
This article is cited by
-
18F-FDG PET/CT metabolic parameters are correlated with clinical features and valuable in clinical stratification management in patients of castleman disease
Cancer Imaging (2025)
-
Over a decade of successful treatment with tocilizumab in a maintenance hemodialysis patient for Castleman disease: a case report and literature review
Renal Replacement Therapy (2025)
-
Structural basis for ligand promiscuity and high signaling activity of Kaposi’s Sarcoma-associated Herpesvirus-encoded GPCR
Nature Communications (2025)
-
Unicentric Castleman disease following POEMS syndrome remission
Journal of Hematopathology (2025)
-
The morphological spectrum of Castleman disease and related disorders: a report from the Lymphoma Workshop of the 22nd Meeting of the European Association of Hematopathology
Virchows Archiv (2025)


