HIV-associated multicentric Castleman\'s disease

June 13, 2017 | Autor: Mark Bower | Categoria: HIV, Humans, American
Share Embed


Descrição do Produto

HIV-associated multicentric Castleman’s disease Justin Stebbing,1* Liron Pantanowitz,2 Farshid Dayyani,3 Ryan J. Sullivan,3 Mark Bower,4,5 and Bruce J. Dezube3

Multicentric Castleman’s disease (MCD), a relatively rare lymphoproliferative disorder that presents with heterogenous symptoms including fevers, anemia, and multifocal lymphadenopathy, is today most commonly observed in individuals infected with human immunodeficiency virus type-1 (HIV). In such individuals, a lymph node biopsy typically identifies cells that stain for Kaposi’s sarcoma-associated herpesvirus proteins, and most HIV-associated MCD features can be attributed to the presence of this c-herpesvirus. Surgery and antiviral therapies including highly active antiretroviral therapy, interferon-a, foscarnet, ganciclovir, and antibodies to interleukin-6 have proved largely ineffective, and chemotherapy in HIV positive individuals is complicated by limited efficacy and pronounced toxicity. While no randomized trials have been performed, more recently the use of the anti-CD20 monoclonal antibody rituximab in large single center cohorts has been associated with prolonged remissions, radiologic responses, as well as hematologic and serum chemistry normalization of the inflammatory picture observed, at the expense of B cell depletion and flare of Kaposi’s sarcoma. MCD represents a model of disease at the interplay between tumor biology, infecC 2008 Wiley-Liss, Inc. tion, and immunology. Am. J. Hematol. 83:498–503, 2008. V Introduction Although there is a trend to eradicate case reports and anecdotes from the medical literature, it is worth recalling an initial description of a particular individual from the Massachusetts General Hospital in 1954 with hyperplastic mediastinal nodes [1]. Two years later, Benjamin Castleman described 13 similar cases of localized, asymptomatic mediastinal masses, and concomitant lymph node hyperplasia resembling thymoma [2]. Thence called Castleman’s disease, is a rare lymphoproliferative disorder whose pathogenesis appears to be related to an aggressive immune response, in many cases against its causative agent Kaposi’s sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus-8 [3]. There are two main forms: unicentric Castleman’s disease is typically an asymptomatic localized condition cured by surgical resection; in contrast multicentric Castleman’s disease (MCD) is usually an aggressive condition associated with systemic symptoms, diffuse peripheral lymphadenopathy, an association with KSHV infection, and an apparent predilection for human immunodeficiency virus (HIV)positive individuals in which preexisting immunosuppression provides the environment for KSHV lytic replication and subsequent cellular transformation. No published literature has convincingly identified the prevalence of MCD, and no studies have compared HIV positive and seronegative cases. However, with the emergence of the HIV pandemic [4], there has been a resurgence of interest secondary to an increasing number of reports of HIV-associated MCD [5]. This review seeks to highlight and update the clinical manifestations, pathobiology, and management issues specific to HIV-associated MCD. Clinical Manifestations Patients with multicentric Castleman’s disease (MCD) present with a clinical picture consistent with an inflammatory process although features at presentation appear to show no difference between human immunodeficiency virus (HIV)-seropositive and negative cases. In a retrospective study of 20 HIV-seropositive individuals with MCD, the main symptoms at presentation were fever, peripheral lymphade-

nopathy, hepatosplenomegaly, weight loss, respiratory symptoms, and edema [5]. In addition, every patient had anemia, an increased level of C-reactive protein, polyclonal hypergammaglobulinemia, hypoalbuminemia, and seven were also pancytopenic. Gender data reported in nine case series and therapeutic trials show that 64 of the 92 affected individuals (70%) were male, although this may reflect the general features of the cohorts studied [6–14]. The pathologic lesions of MCD include lymphadenopathy and/or localized extranodal lymphoid tumors with involved lymph nodes grossly measuring 3–7 cm in size [15]. Significant fibrosis, calcification, and even ossification can be seen [16,17], and bone marrow biopsies contain either focal infiltrates of plasma cells [18,19] or plasmablasts within lymphoid follicles and the interstitium [20]. In the lung, a lymphocytic interstitial pneumonia may occur, with thickened alveoli containing plasma cells admixed with fibroblasts [21,22]. Cases may also be complicated by the coexistence or subsequent development of non-Hodgkin lymphoma [23,24], follicular dendritic cell sarcoma [25], Hodgkin lymphoma [26], and another Kaposi’s sarcomaassociated herpesvirus (KSHV)-driven neoplasm primary effusion lymphoma [27].

This project was funded in part by a grant from the AIDS Malignancy Consortium of the National Cancer Institute to BJD. 1 Department of Medical Oncology, Imperial College School of Science, Technology and Medicine, The Hammersmith Hospitals NHS Trust, London, United Kingdom; 2Department of Pathology, Baystate Medical Center, Tufts University School of Medicine, Springfield, Massachusetts; 3Department of Medicine, Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; 4Department of Oncology, The Chelsea and Westminster Hospital, London, United Kingdom; 5 Department of HIV Medicine, The Chelsea and Westminster Hospital, London, United Kingdom

*Correspondence to: Bruce Dezube, MD, Beth Israel Deaconess Medical Center, 330 Brookline Ave, MASCO 414, Boston, MA 02215. E-mail: [email protected] Received for publication 2 December 2007; Accepted 5 December 2007 Am. J. Hematol. 83:498–503, 2008. Published online 7 February 2008 in Wiley InterScience (www.interscience.wiley. com). DOI: 10.1002/ajh.21137

C 2008 Wiley-Liss, Inc. V

American Journal of Hematology

498

http://www3.interscience.wiley.com/cgi-bin/jhome/35105

Given the association with KSHV, it is not surprising that Kaposi’s sarcoma has been concomitantly noted in 13% of patients with unicentric disease and 75% of individuals with MCD [6]. MCD can also be complicated by neurologic manifestations including polyneuropathy and myesthenia gravis [28–30], either of which may be part of a pentad, in concert with organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes, known as the POEMS syndrome [29,31]; CNS involvement by MCD lesions has also been described [32]. The first step toward successfully making the diagnosis of MCD is to consider it in high risk patients, especially if they present with associated conditions (e.g., HIV-infection, Kaposi’s sarcoma, primary effusion lymphoma). Ultimately, the diagnosis needs to be established histologically from procured tissue, usually as an excised lymph node. Once the diagnosis has been established, further workup including laboratory tests and imaging is warranted. Laboratory studies should include where available, testing for KSHV DNA in serum or from peripheral blood mononuclear cells by real time PCR. A study of 31 cases of MCD with abdominal localization showed that the size of the affected lymph nodes determined the appearance on imaging: nodes smaller than 5 cm in diameter showed a homogenous enhancement, and nodes greater than 5 cm were dyshomogenous. In an attempt to distinguish NHL from MCD, Chim et al. studied gallium (67 Ga) uptake in three patients: while lymphomas were shown to readily take up 67 Ga, in all three patients with MCD 67 Ga uptake was absent. It remains to be determined how well negative 67 Ga uptake will distinguish MCD from other nonmalignant lymphadenopathy [33]. Imaging, including use of 18-FDG PET, may be a valuable tool for follow-up and diagnosing relapse after remission with systemic therapy [34]. Pathogenesis The manifestations of MCD are in part due to cytokine dysregulation including increased levels of interleukin-6 (IL6) [35–37], which acts as a B-cell stimulatory factor, mediating B-cell differentiation as well as promoting the growth of B-cell malignancies [38–40]. C/EBP b is a transcriptional regulator of IL-6, and mice that lack this gene overexpress IL-6 and develop a condition similar to MCD [41]; inactivation of IL-6 in these knockout mice prevents the development of this murine Castleman’s like disease [42]. KSHV induces IL-6 production in infected cells [43], and KSHV itself encodes an early lytic pirated homologue of IL-6 [44– 49] (vIL-6). Although there are subtle differences in the receptor activating signaling complex between its human and viral homologues [50–52], KSHV encoded vIL-6 stimulates the known hIL-6-induced signaling pathways via the shared cytokine signaling receptor gp130 coupled to the endogenous JAK/STAT pathway. In mice, these data are supported by evidence that recombinant vIL-6 induces a marked plasmacytosis similar to that found in MCD, as well as accelerating hematopoesis and inducing vascular endothelial growth factor, a proangiogenic cytokine [53]. Additionally, in MCD, high KSHV viral replication and high levels of IL-6, IL-10, and C-reactive protein are associated with a more aggressive disease course, suggesting that these cytokines may be involved in the pathogenesis of this disease [54]. Finally, IL-6 levels have been shown to decrease concomitantly with KSHV levels during therapy [55]. The clonality of MCD and its progression to lymphoma is similarly influenced by KSHV. KSHV has been detected in the large mantle zone plasmablasts of MCD [56]. These plasmablasts express high levels of lambda chain restricted IgM; however, in the interfollicular region mature B cells are KSHV negative, IgM negative, and polytypic. These KSHV

American Journal of Hematology

positive, IgM lambda restricted plasmablasts are often isolated cells, but they may coalesce into microscopic aggregates known as microlymphomas and in some cases develop into frank plasmablastic lymphomas. The clonality of plasmablasts in 13 cases of MCD including eight with microlymphomas and two with plasmablastic lymphomas were evaluated by immunoglobulin (Ig) gene rearrangement studies. These studies demonstrated that the KSHV positive plasmablasts were polyclonal in MCD-involved lymphoid tissue and in six of eight microlymphomas, but monoclonal in two individuals with microlymphoma and plasmablastic lymphoma [14]. Moreover, the absence of somatic Ig gene rearrangements suggests that the KSHV positive plasmablasts are derived from naı¨ve B cells. KSHVencoded vIL-6 was only detected in 10–15% of KSHV positive plasmablasts; however, the hIL-6 receptor was expressed by all KSHV positive plasmablasts. It can be hypothesized that activation of the IL-6 signaling pathway by KSHV vIL-6 may transform naı¨ve B cells into plasmablasts and lead to the lymphoproliferative diseases associated with this virus including MCD. Pathologic Characteristics There are two main pathologic types of MCD, hyalinevascular, and plasma-cell types according to the histologic features of the affected lymph nodes. The latter type is more likely to be encountered in an excisional biopsy from an HIV-positive patient. The hyaline-vascular type is rarely seen in HIV-infected individuals, unless it coexists with the plasma cell type (i.e., mixed variant). In the hyaline-vascular variant, lymph node architecture is altered by an increased number of abnormal lymphoid follicles. Often germinal centers appear to be transfixed by radially penetrating capillaries surrounded by a broad mantle of concentrically arranged lymphocytes, so called ‘‘onion skin’’ layering giving the follicle a target appearance (Fig. 1A and 1B). The interfollicular region also contains numerous proliferating blood vessels, and such hypervascularity explains the occasional significant bleeding during surgery [57]. Stromal cell proliferations, such as angiomyoid proliferative lesions [58] and fibrous pseudotumors [59], may occasionally supervene. These stroma-rich variants likely arise from proliferating interfollicular actin-positive fibroblastic reticulum cells (myoid cells) and dendritic cells [60]. Lymphoid follicles in the plasma cell variant tend to be more hyperplastic, with larger and more active germinal centers that are surrounded by a narrower mantle of mature lymphocytes (Fig. 1C). These follicles usually have poorly defined borders, although hyalinized follicles may occasionally be present. The overall size of the lymphoid follicles, however, is similar to that seen in the hyaline-vascular type [61]. Although the interfollicular areas and medulla are similarly vascular, referred to as being ‘‘Kaposilike’’ by some authors [62], they are predominantly expanded by large sheets of plasma cells. The plasmablastic variant, the most common type observed during HIV infection, is characterized by increased numbers of medium-sized to large plasmablasts which morphologically resemble immunoblasts. They are characterized by a moderate amount of amphophilic cytoplasm and a large vesicular nucleus with 1–2 prominent nucleoli. Plasmablasts can comprise up to 50% of the follicular mantle zone in some follicles, and may also colonize the germinal centers [14]. They may even coalesce to form ‘‘microlymphomas’’ and develop into frank lymphoma. Immunohistochemical staining with a monoclonal antibody directed against the latency-associated nuclear antigen of KSHV often shows nuclear staining of B cells (Fig. 1D), located in 10–30% of lymphocytes in the mantle

499

Figure 1. A: Small target-shaped lymphoid follicle with an involuted germinal center and mantle comprised of concentric layers of small lymphocytes (H&E stain). B: A germinal center is seen transfixed by a penetrating blood vessel (so-called lollipop structure) (H&E stain). C: A hyperplastic lymphoid follicle seen in a case of plasma cell variant Castleman disease (H&E stain). D: Several lymphocytes, including plasmablasts, within the mantle zone of this lymphoid follicle are KSHV1 in a case of HIV-associated plasmablastic variant Castleman’s disease (LNA-1 immunohistochemical stain). [Color figure can be viewed in the online issue, which is available at www.interscience.wiley.com.]

zone [56,63], in germinal center macrophages [64], as well as in endothelial cells and subcapsular spindle cell proliferations [63]. The highest copy number of KSHV appears to be present within subcapsular spindle cells [63]. In the plasmablastic variant, KSHV positive cells are mainly plasmablasts [14,65]. EBV can occasionally also be detected in some cases by means of immunohistochemistry (LMP-1), in situ hybridization studies (EBER) and molecular techniques [66,67]. Treatment There has never been a randomized study in any form of MCD, and the best evidence in HIV-positive patients has been derived from two recent clinical cohorts using rituximab [68,69]. Surgery Surgery has a limited role in MCD although in unicentric Castleman’s disease complete removal of the mediastinal lesions is curative. Splenectomy, in addition to establishing the histological diagnosis, may have a therapeutic benefit as a debulking procedure. Some of the hematological

500

sequelae such as thrombocytopenia and anemia may in part be due to splenomegaly. Following splenectomy, there is often resolution of the constitutive symptoms, but this is typically short lived, and 1–3 months later symptoms tend to relapse, and an additional form of therapy is required [5]. Chemotherapy For immunocompetent patients, chemotherapy regimens for MCD are based on lymphoma schedules such as CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone). In the pre-HAART era, these schedules were associated with marked toxicity in HIV-positive individuals, and consequently, other schedules were developed. In the largest published study from Paris of 20 HIV-seropositive patients, the partial response rate was 100% (nine out of nine patients) with single agent vinblastine; however, only four patients remained stable with maintenance therapy (4–6 mg/2 weeks). Five patients relapsed and required combination chemotherapy, which was ABV (doxorubicin, bleomycin, and vincristine) or partial splenectomy. Four patients received up-front ABV, and three had a partial

American Journal of Hematology

response. In three patients, intermittent treatment with cyclophosphamide achieved a partial response [5]. Two HIVpositive patients were treated with oral etoposide for MCD and had remissions of 1.5 and 6 months, respectively, although the former patient had previous chemotherapy and ganciclovir [70]. Although there are few published data on which to base treatment strategies, in many centers combination chemotherapy is used initially to induce remission in aggressive forms of MCD. This may be followed by single agent chemotherapy, such as vinblastine or etoposide, as maintenance. HAART The introduction of highly active antiretroviral therapy (HAART) has been associated with a reduction in the incidence of many HIV-associated malignancies including Kaposi’s sarcoma [71]. Furthermore, HAART leads to a decrease in KSHV viral replication and is associated with the resolution of individual KS lesions and prolonged time to progression [72,73]. The effect of HAART has been described in seven patients with HIV-associated MCD [74]. Six of these patients were treated with chemotherapy and responded to it, and immune reconstitution was observed in five patients. However, patients in this study continued to require long-term chemotherapy to prevent further episodes of MCD. The mean survival was 48 months, which was longer than described in the pre-HAART era when most individuals succumbed to opportunistic infections related to their HIV infection. Interestingly, in an early case series of three HIV-associated MCD patients treated with HAART, the development of aggressive MCD was thought to be part of the immune reconstitution disease spectrum associated with the starting of HAART [75]. Unfortunately, all three patients died, indicating the life threatening nature of this disease, particularly in the setting of immune reconstitution. Immunotherapy Interferon-a has been administered either alone or in combination with HAART or chemotherapy for patients with HIV-associated MCD, both to induce remission and as maintenance therapy [5,76,77]. Mechanistically, this is thought to have both an antiproliferative effect by directly binding to cell surface receptors, and an antiviral effect by inhibiting viral replication and increasing natural killer cell activity via upregulation of the major histocompatibility class I expression of KSHV infected cells [78]. In combination with vinblastine and splenectomy, long-term remission was seen in two-thirds of patients [5]. In a case report, a patient was initially treated with antiviral therapy and splenectomy, followed by chemotherapy to induce remission. When chemotherapy failed to achieve sustained remission, interferon-a therapy was started leading to remission for over a year [76]. Another report of treatment of MCD with HAART and low dose interferon-a alone demonstrated a sustained remission of 24 months [79]. In HIV-seronegative individuals with MCD, the use of anti-IL-6R monoclonal antibodies (altizumab) [9] and separately the use of thalidomide [80,81] in small series has met with some success, but their efficacy and toxicity in HIV-associated MCD has not been defined. Anti-KSHV therapy The effect of specific anti-herpesvirus therapy to reduce the KSHV viral replication and alleviate disease has been examined in KSHV-associated diseases in the HIV setting. Kaposi’s sarcoma incidence was reduced when prophylactic ganciclovir or foscarnet was used to prevent CMV retinitis [82,83], and antiviral treatment which has led to a clinical improvement has been shown to reduce KSHV viral

American Journal of Hematology

replication in patients with Kaposi’s sarcoma [84], primary effusion lymphoma, and hemophagocytic syndrome [85]. In a series of three patients treated with ganciclovir, there was a reduction in the frequency of acute symptoms of MCD for two patients treated with oral and intravenous ganciclovir [7]. For the third patient, who was in the intensive care unit, there was resolution of pulmonary and renal failure with intravenous ganciclovir. All the patients had a reduction in KSHV viral replication with ganciclovir therapy, accompanying the resolution of their symptoms. However, the use of foscarnet and cidofovir antiviral therapy was ineffective in a HIV-negative MCD patient with proven KSHV viremia, and treatment with corticosteriods in combination with chlorambucil chemotherapy was required to achieve a clinical response [86]. While there are few data on valganciclovir [87], a short course (e.g., 3 months) of this antiviral is often used following rituximab therapy by clinicians treating HIV-associated MCD; KHSV replication may be monitored during the valganciclovir treatment period. Rituximab The use of the anti-CD20 monoclonal antibody rituximab routinely prescribed as therapy for B cell lymphomas and autoimmune diseases, to target KSHV infected plasmablasts in MCD is a novel and potentially beneficial approach to the treatment of this disease. Before 2007, it has been the subject of case reports and very small clinical series, totaling 12 patients. These patients were often pretreated with chemotherapy and follow-up was brief [13,55,88–91,92], however, most patients (10 of 12) experienced a complete response. More recently, the efficacy and safety of four weekly infusions of 375 mg/m2 of rituximab in 21 consecutive patients with previously untreated plasmablastic HIV-associated has been investigated [69]. Apart from one individual who died from multiorgan failure within 2 weeks of receiving rituximab, all achieved clinical remission of symptoms, hematological and serum chemistry normalization, and 70% achieved a radiological response. The overall survival and disease free survival at 2 years were 95% and 79%, respectively, with no grade 3 or 4 toxicities. In three patients who relapsed, retreatment with rituximab was successful [93]. These data corroborate the benefit seen in the aforementioned case reports and demonstrate that rituximab therapy results in an impressive clinical, biochemical, and radiological sustained response in HIV-related MCD (see Fig. 2). The main adverse event seen in these patients is reactivation of Kaposi’s sarcoma, which is intriguing and may be due to a rapid B cell depletion that is observed during rituximab therapy, or an immune reconstitution inflammatory syndrome to hitherto latent antigens [94]. Rituximab therapy was associated with a decline in KSHV levels initially and at the successful treatment of relapse. In a prospective study of 24 individuals with chemotherapy-dependent HIV-associated MCD, the ANRS 117 CastlemaB Trial [68], use of the same schedule of rituximab was associated with sustained remission off treatment at day 60 (the primary end point) in 22 patients (92%). Conclusions While the optimal treatment for HIV-associated MCD is debatable, we personally favor initiating treatment with upfront rituximab. Corticosteroids and/or chemotherapy may be given depending on the situation (disease burden, clinical presentation). HAART is necessary to treat the underlying HIV infection. For those patients who are antiretroviral naı¨ve at the time of MCD diagnosis, initiation of HAART may be delayed until a course of rituximab has been given. A short course of valganciclovir (e.g., 3 months) may be useful to control KSHV replication until the patient’s immune system has been partially reconstituted

501

Figure 2. A: CT scan pre- (top) and post- (bottom) 4 infusions of weekly rituximab demonstrating resolution of splenomegaly and lymphadenopathy (white arrow). B: FDG-PET scan performed at relapse. The patient had been treated 14 months previously with a splenectomy and rituximab and represented with fevers, anemia and lymphadenopathy. The PET scan shows increased activity in axillary, supraclavicular, paratracheal, coeliac axis, iliac and inguinal lymph nodes and absence of the spleen. The patient responded to retreatment with rituximab.

by HAART. The challenge for the scientific community that investigates and treats MCD is to further the understanding of the disease process and to optimize and enhance the therapeutic strategies available to patients. This also presents opportunities to expand our understanding and treatment of viral oncogenesis. References 1. Castleman B, Towne VW. Case records of the Massachusetts general hospital: Case No. 40231. N Engl J Med 1954;250:1001–1005. 2. Castleman B, Iverson L, Menendez VP. Localized mediastinal lymphnode hyperplasia resembling thymoma. Cancer 1956;9:822–830. 3. Soulier J, Grollet L, Oksenhendler E, et al. Kaposi’s sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman’s disease. Blood 1995; 86:1276–1280.

502

4. Stebbing J, Bower M. What can oncologists learn from HIV? Lancet Oncol 2003;4:438–445. 5. Oksenhendler E, Duarte M, Soulier J, et al. Multicentric Castleman’s disease in HIV infection: A clinical and pathological study of 20 patients. AIDS 1996; 10:61–67. 6. Bowne WB, Lewis JJ, Filippa DA, et al. The management of unicentric and multicentric Castleman’s disease: A report of 16 cases and a review of the literature. Cancer 1999;85:706–717. 7. Casper C, Nichols WG, Huang ML, et al. Remission of HHV-8 and HIV-associated multicentric Castleman disease with ganciclovir treatment. Blood 2004; 103:1632–1634. 8. Nishimoto N, Sasai M, Shima Y, et al. Improvement in Castleman’s disease by humanized anti-interleukin-6 receptor antibody therapy. Blood 2000;95:56–61. 9. Nishimoto N, Kanakura Y, Aozasa K, et al. Humanized anti-interleukin-6 receptor antibody treatment of multicentric Castleman disease. Blood 2005; 106:2627–2632. 10. Colleoni GW, Duarte LC, Kerbauy FR, et al. 2-Chloro-deoxyadenosine induces durable complete remission in Castleman’s disease but may accelerate its transformation to non-Hodgkin’s lymphoma. Acta Oncol 2003;42:784–787. 11. Soulier J, Grollet L, Oksenhendler E, et al. Molecular analysis of clonality in Castleman’s disease. Blood 1995;86:1131–1138. 12. Ide M, Kawachi Y, Izumi Y, et al. Long-term remission in HIV-negative patients with multicentric Castleman’s disease using rituximab. Eur J Haematol 2006;76:119–123. 13. Neuville S, Agbalika F, Rabian C, et al. Failure of rituximab in human immunodeficiency virus-associated multicentric Castleman disease. Am J Hematol 2005;79:337–339. 14. Dupin N, Diss TL, Kellam P, et al. HHV-8 is associated with a plasmablastic variant of Castleman disease that is linked to HHV-8-positive plasmablastic lymphoma. Blood 2000;95(4):1406–1412. 15. Ioachim HL, Ratech H. Ioachim’s Lymph Node Pathology. Philadelphia: Lippincott, Williams & Wilkins; 2002. pp 246–253. 16. Ota T, Mitsuyoshi A, Zaima M, et al. Visualization of central stellate fibrosis in hyaline vascular type Castleman’s disease. Br J Radiol 1997;70:1060–1062. 17. Goetze O, Banasch M, Junker K, et al. Unicentric Castleman’s disease of the pancreas with massive central calcification. World J Gastroenterol 2005;11: 6725–6727. 18. Kreft A, Weber A, Springer E, et al. Bone marrow findings in multicentric Castleman disease in HIV-negative patients. Am J Surg Pathol 2007;31:398–402. 19. Molina T, Brouland JP, Bigorgne C, et al. [Pseudo-myelomatous plasmacytosis of the bone marrow in a multicentric Castleman’s disease]. Ann Pathol 1996;16:133–136. 20. Bacon CM, Miller RF, Noursadeghi M, et al. Pathology of bone marrow in human herpes virus-8 (HHV8)-associated multicentric Castleman disease. Br J Haematol 2004;127:585–591. 21. Frizzera G. Atypical lymphoproliferative disorders. In: Neoplastic Hematopathology. Knowles DM, editor. Philadelphia: Lippincott, Williams & Wilkins; 2001. pp 569–622. 22. Johkoh T, Muller NL, Ichikado K, et al. Intrathoracic multicentric Castleman disease: CT findings in 12 patients. Radiology 1998;209:477–481. 23. Kojima M, Nakamura S, Shimizu K, et al. Nodal marginal zone B-cell lymphoma resembling plasmacytoma arising from a plasma cell variant of localized Castleman’s disease: A case report. APMIS 2002;110(7/8):523–527. 24. Venizelos I, Tamiolakis D, Simopoulos C, et al. Diffuse large B-cell lymphoma arising from a multicentric mixed variant of Castleman’s disease. Indian J Cancer 2004;41:135–137. 25. Chan JK, Tsang WY, Ng CS. Follicular dendritic cell tumor and vascular neoplasm complicating hyaline-vascular Castleman’s disease. Am J Surg Pathol 1994;18:517–525. 26. Zarate-Osorno A, Medeiros LJ, Danon AD, et al. Hodgkin’s disease with coexistent Castleman-like histologic features. A report of three cases. Arch Pathol Lab Med 1994;118:270–274. 27. Stebbing J, Wilder N, Ariad S, et al. Lack of intra-patient strain variability during infection with Kaposi’s sarcoma-associated herpesvirus. Am J Hematol 2001;68:133–134. 28. Day JR, Bew D, Ali M, et al. Castleman’s disease associated with myasthenia gravis. Ann Thorac Surg 2003;75:1648–1650. 29. Papo T, Soubrier M, Marcelin AG, et al. Human herpesvirus 8 infection, Castleman’s disease and POEMS syndrome. Br J Haematol 1999;104:932–933. 30. Pasaoglu I, Dogan R, Topcu M, et al. Multicentric angiofollicular lymph-node hyperplasia associated with myasthenia gravis. Thorac Cardiovasc Surg 1994;42:253–256. 31. Waterston A, Bower M. Fifty years of multicentric Castleman’s disease. Acta Oncol 2004;43:698–704. 32. Ribeiro LT, Simao GN, Matos AL, et al. Intracranial Castleman’s disease presenting as hypopituitarism. Neuroradiology 2004;46:830–833. 33. Chim CS, Choi FP, Ooi GC, et al. Absence of gallium uptake in multicentric Castleman’s disease of plasma cell type. Haematologica 2001;86:442–443. 34. Enomoto K, Nakamichi I, Hamada K, et al. Unicentric and multicentric Castleman’s disease. Br J Radiol 2007;80:e24–e26. 35. Aoki Y, Tosato G, Fonville TW, et al. Serum viral interleukin-6 in AIDS-related multicentric Castleman disease. Blood 2001;97:2526–2527. 36. Choi J, Means RE, Damania B, et al. Molecular piracy of Kaposi’s sarcoma associated herpesvirus. Cytokine Growth Factor Rev 2001;12(2/3):245–257. 37. Said J. Kaposi’s sarcoma-associated herpesvirus (KSHV): A new viral pathogen associated with Kaposi’s sarcoma, primary effusion lymphoma, and multicentric Castleman’s disease. West J Med 1997;167:37–38.

American Journal of Hematology

38. Du MQ, Liu H, Diss TC, et al. Kaposi sarcoma-associated herpesvirus infects monotypic (IgM k) but polyclonal naive B cells in Castleman disease and associated lymphoproliferative disorders. Blood 2001;97:2130–2136. 39. van Kooten C, Rensink I, Aarden L, et al. Effect of IL-4 and IL-6 on the proliferation and differentiation of B-chronic lymphocytic leukemia cells. Leukemia 1993;7:618–624. 40. Yokoi T, Miyawaki T, Yachie A, et al. Epstein-Barr virus-immortalized B cells produce IL-6 as an autocrine growth factor. Immunology 1990;70:100–105. 41. Brandt SJ, Bodine DM, Dunbar CE, et al. Dysregulated interleukin 6 expression produces a syndrome resembling Castleman’s disease in mice. J Clin Invest 1990;86:592–599. 42. Screpanti I, Musiani P, Bellavia D, et al. Inactivation of the IL-6 gene prevents development of multicentric Castleman’s disease in C/EBP b-deficient mice. J Exp Med 1996;184:1561–1566. 43. Deng H, Song MJ, Chu JT, et al. Transcriptional regulation of the interleukin-6 gene of human herpesvirus 8 (Kaposi’s sarcoma-associated herpesvirus). J Virol 2002;76:8252–8264. 44. An J, Lichtenstein AK, Brent G, et al. 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 2002;99:649–654. 45. Hsu SM, Waldron JA, Xie SS, et al. Expression of interleukin-6 in Castleman’s disease. Hum Pathol 1993;24:833–839. 46. Oksenhendler E, Boulanger E, Galicier L, et al. High incidence of Kaposi sarcoma-associated herpesvirus-related non-Hodgkin lymphoma in patients with HIV infection and multicentric Castleman disease. Blood 2002;99:2331–2336. 47. Russo JJ, Bohenzky RA, Chien MC, et al. Nucleotide sequence of the Kaposi sarcoma-associated herpesvirus (HHV8). Proc Natl Acad Sci USA 1996; 93:14862–14867. 48. Song J, Ohkura T, Sugimoto M, et al. Human interleukin-6 induces human herpesvirus-8 replication in a body cavity-based lymphoma cell line. J Med Virol 2002;68:404–411. 49. Staskus KA, Sun R, Miller G, 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 1999;73:4181–4187. 50. Boulanger MJ, Chow DC, Brevnova E, et al. Molecular mechanisms for viral mimicry of a human cytokine: Activation of gp130 by HHV-8 interleukin-6. J Mol Biol 2004;335:641–654. 51. Li H, Wang H, Nicholas J. Detection of direct binding of human herpesvirus 8-encoded interleukin-6 (vIL-6) to both gp130 and IL-6 receptor (IL-6R) and identification of amino acid residues of vIL-6 important for IL-6R-dependent and -independent signaling. J Virol 2001;75:3325–3334. 52. Molden J, Chang Y, You Y, et al. A Kaposi’s sarcoma-associated herpesvirusencoded cytokine homolog (vIL-6) activates signaling through the shared gp130 receptor subunit. J Biol Chem 1997;272:19625–19631. 53. Aoki Y, Jaffe ES, Chang Y, et al. Angiogenesis and hematopoiesis induced by Kaposi’s sarcoma-associated herpesvirus-encoded interleukin-6. Blood 1999; 93:4034–4043. 54. Oksenhendler E, Carcelain G, Aoki Y, 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 2000;96:2069–2073. 55. Newsom-Davis T, Bower M, Wildfire A, et al. Resolution of AIDS-related Castleman’s disease with anti-CD20 monoclonal antibodies is associated with declining IL-6 and TNF-a levels. Leuk Lymphoma 2004;45:1939–1941. 56. Dupin N, Fisher C, Kellam P, 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 1999;96:4546– 4551. 57. Pereira TC, Landreneau R, Nathan G, et al. Pathologic quiz case. Large posterior mediastinal mass in a young woman. Pathologic diagnosis: Localized hyaline-vascular-type Castleman disease (angiofollicular lymphoid hyperplasia). Arch Pathol Lab Med 2001;125:964–967. 58. Izumi M, Mochizuki M, Kuroda M, et al. Angiomyoid proliferative lesion: An unusual stroma-rich variant of Castleman’s disease of hyaline-vascular type. Virchows Arch 2002;441:400–405. 59. Dargent JL, Delplace J, Roufosse C, et al. Development of a calcifying fibrous pseudotumour within a lesion of Castleman disease, hyaline-vascular subtype. J Clin Pathol 1999;52:547–549. 60. Danon AD, Krishnan J, Frizzera G. Morpho-immunophenotypic diversity of Castleman’s disease, hyaline-vascular type: With emphasis on a stroma-rich variant and a new pathogenetic hypothesis. Virchows Arch A Pathol Anat Histopathol 1993;423:369–382. 61. Menke DM, Tiemann M, Camoriano JK, et al. Diagnosis of Castleman’s disease by identification of an immunophenotypically aberrant population of mantle zone B lymphocytes in paraffin-embedded lymph node biopsies. Am J Clin Pathol 1996;105:268–276. 62. Kim JE, Kim CJ, Park IA, et al. Clinicopathologic study of Castleman’s disease in Korea. J Korean Med Sci 2000;15:393–398. 63. O’Leary J, Kennedy M, Howells D, et al. Cellular localisation of HHV-8 in Castleman’s disease: Is there a link with lymph node vascularity? Mol Pathol 2000;53:69–76. 64. Valmary S, Richard P, Brousset P. Frequent detection of Kaposi’s sarcoma herpesvirus in germinal centre macrophages from AIDS-related multicentric Castleman’s disease. AIDS 2005;19:1229–1231.

American Journal of Hematology

65. Scadden DT, Muse VV, Hasserjian RP. Case records of the Massachusetts general hospital. Case 30-2006. A 41-year-old man with dyspnea, fever, and lymphadenopathy. N Engl J Med 2006;355:1358–1368. 66. Hanson CA, Frizzera G, Patton DF, et al. Clonal rearrangement for immunoglobulin and T-cell receptor genes in systemic Castleman’s disease. Association with Epstein-Barr virus. Am J Pathol 1988;131:84–91. 67. Murray PG, Deacon E, Young LS, et al. Localization of Epstein-Barr virus in Castleman’s disease by in situ hybridization and immunohistochemistry. Hematol Pathol 1995;9:17–26. 68. Gerard L, Berezne A, Galicier 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 2007;25:3350–3356. 69. Bower M, Powles T, Williams S, Davis TN, Atkins M, Montoto S, Orkin C, Webb A, Fisher M, Nelson M, Gazzard B, Stebbing J, Kelleher P. Rituximab in HIV-associated multicentric Castleman disease. Ann Intern Med 2007; 147:836–839. 70. Scott D, Cabral L, Harrington WJ Jr. Treatment of HIV-associated multicentric Castleman’s disease with oral etoposide. Am J Hematol 2001;66:148–150. 71. Palella FJ Jr, Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998;338:853–860. 72. Bower M, Fox P, Fife K, et al. Highly active anti-retroviral therapy (HAART) prolongs time to treatment failure in Kaposi’s sarcoma. AIDS 1999;13:2105–2111. 73. Portsmouth S, Stebbing J, Gill J, et al. A comparison of regimens based on non-nucleoside reverse transcriptase inhibitors or protease inhibitors in preventing Kaposi’s sarcoma. AIDS 2003;17:F17–F22. 74. Aaron L, Lidove O, Yousry C, et al. Human herpesvirus 8-positive Castleman disease in human immunodeficiency virus-infected patients: The impact of highly active antiretroviral therapy. Clin Infect Dis 2002;35:880–882. 75. Zietz C, Bogner JR, Goebel FD, et al. An unusual cluster of cases of Castleman’s disease during highly active antiretroviral therapy for AIDS. N Engl J Med 1999;340:1923–1924. 76. Nord JA, Karter D. Low dose interferon-alpha therapy for HIV-associated multicentric Castleman’s disease. Int J STD AIDS 2003;14:61–62. 77. Strohal R, Tschachler E, Breyer S, et al. Reactivation of Behcet’s disease in the course of multicentric HHV8-positive Castleman’s disease: Long-term complete remission by a combined chemo/radiation and interferon-a therapy regimen. Br J Haematol 1998;103:788–790. 78. Lebbe C, Agbalika F, de Cremoux P, et al. Detection of human herpesvirus 8 and human T-cell lymphotropic virus type 1 sequences in Kaposi sarcoma. Arch Dermatol 1997;133:25–30. 79. Kumari P, Schechter GP, Saini N, et al. Successful treatment of human immunodeficiency virus-related Castleman’s disease with interferon-a. Clin Infect Dis 2000;31:602–604. 80. Jung CP, Emmerich B, Goebel FD, et al. Successful treatment of a patient with HIV-associated multicentric Castleman disease (MCD) with thalidomide. Am J Hematol 2004;75:176–177. 81. Lee FC, Merchant SH. Alleviation of systemic manifestations of multicentric Castleman’s disease by thalidomide. Am J Hematol 2003;73:48–53. 82. Martin DF, Kuppermann BD, Wolitz RA, et al. Oral ganciclovir for patients with cytomegalovirus retinitis treated with a ganciclovir implant. Roche Ganciclovir Study Group. N Engl J Med 1999;340:1063–1070. 83. Mazzi R, Parisi SG, Sarmati L, et al. Efficacy of cidofovir on human herpesvirus 8 viraemia and Kaposi’s sarcoma progression in two patients with AIDS. AIDS 2001;15:2061–2062. 84. Low P, Neipel F, Rascu A, et al. Suppression of HHV-8 viremia by foscarnet in an HIV-infected patient with Kaposi’s sarcoma and HHV-8 associated hemophagocytic syndrome. Eur J Med Res 1998;3:461–464. 85. Luppi M, Barozzi P, Rasini V, et al. Severe pancytopenia and hemophagocytosis after HHV-8 primary infection in a renal transplant patient successfully treated with foscarnet. Transplantation 2002;74:131–132. 86. Senanayake S, Kelly J, Lloyd A, et al. Multicentric Castleman’s disease treated with antivirals and immunosuppressants. J Med Virol 2003;71:399–403. 87. Valencia ME, Moreno V, Martinez P, et al. [Favorable outcome of Castleman’s disease treated with oral valganciclovir]. Med Clin (Barc) 2005;125:399. 88. Corbellino M, Bestetti G, Scalamogna C, et al. Long-term remission of Kaposi sarcoma-associated herpesvirus-related multicentric Castleman disease with anti-CD20 monoclonal antibody therapy. Blood 2001;98:3473–3475. 89. Marcelin AG, Aaron L, Mateus C, et al. Rituximab therapy for HIV-associated Castleman disease. Blood 2003;102:2786–2788. 90. Marrache F, Larroche C, Memain N, et al. Prolonged remission of HIV-associated multicentric Castelman’s disease with an anti-CD20 monoclonal antibody as primary therapy. AIDS 2003;17:1409–1410. 91. Kofteridis DP, Tzagarakis N, Mixaki I, et al. Multicentric Castleman’s disease: Prolonged remission with anti CD-20 monoclonal antibody in an HIV-infected patient. AIDS 2004;18:585–586. 92. Dayyani F, Pantanowitz L, Sandridge TG, et al. Multicentric Castleman’s disease masquerading as HIV-related lymphoma. Am J Med Sci 2007;334:317– 319. 93. Powles T, Stebbing J, Montoto S, et al. Rituximab as retreatment for rituximab pretreated HIV-associated multicentric Castleman disease. Blood 2007;110: 4132–4133. 94. Bower M, Nelson M, Young AM, et al. Immune reconstitution inflammatory syndrome associated with Kaposi’s sarcoma. J Clin Oncol 2005;23:5224– 5228.

503

Lihat lebih banyak...

Comentários

Copyright © 2017 DADOSPDF Inc.