ISSN 1866-8836
Клеточная терапия и трансплантация

Systemic treatment of chronic GVHD

Michael Schleuning

Zentrum für Blutstammzell- und Knochenmarktransplantation, Wiesbaden, Germany

Correspondence
Prof. Dr. Michael Schleuning, Zentrum für Blutstammzell- und Knochenmarktransplantation, Deutsche Klinik für Diagnostik, Aukammallee 33, 65191 Wiesbaden, Germany
Phone: +49611577169, fax: +49611577313, E-mail: schleuning.kmt@dkd-wiesbaden.de
doi 10.3205/ctt-2009-en-000050.01
Submitted 23 August 2009
Accepted 08 October 2009
Published 16 October 2009

Summary

Severe chronic graft versus host disease (GVHD) is the main factor for late morbidity and mortality in long-term survivors after allogeneic hematopoietic cell transplantation. The only established treatment of chronic graft versus host disease is the use of high dose corticosteroids. However, multiple different treatment approaches have been evaluated mostly in small phase 2 studies. These included calcineurin inhibitors for blocking T-cell activation, classical cytotoxic drugs, like methotrexate or azathioprine, as well as immunomodulatory substances like cytokine inhibitors or thalidomide. More recently novel treatment approaches have been evaluated. These include the use of B-cell antibodies and the tyrosine kinase inhibitor imatinib. Furthermore treatment options beyond mere immunosuppression that aim to induce tolerance are currently under investigation. These include extra-corporal photopheresis and treatment with inhibitors of the mammalian target of rapamycin. This review will discuss these different treatment approaches.

Keywords

chronic GVHD, calcineurin inhibitors, mTOR, extracorporal photopheresis, immunomodulatory drugs, steroids, methotrexate


Introduction

Allogeneic hematopoietic cell transplantation (HCT) is the treatment of choice for high risk hematological malignancies. Its success, however, is hampered by high treatment associated mortality, mostly due to acute and chronic graft versus host disease (GVHD).

Although mild chronic GVHD may improve survival in patients with malignant diseases and usually needs no therapeutic intervention, severe chronic GVHD has devastating effects on multiple organs and significantly contributes to late morbidity and mortality in long-term survivors after allogeneic HCT [1]. The only established treatment of chronic graft versus host disease is the use of high dose corticosteroids. This therapy, however, is associated with multiple adverse reactions including life threatening infections, and moreover not all patients respond to this treatment approach. Therefore alternative treatment options are urgently needed.

Calcineurin inhibitors

In 1988 Sullivan introduced a new alternating-day regimen of prednisone (1 mg/kg every other day) and oral cyclosporine (6 mg/kg every 12 hours every other day) for patients with high-risk extensive chronic GVHD (thrombocytopenia < 100.000/µl) as primary treatment, and achieved with this approach a remarkable 4-year survival of 50%. The overall response rate after 9 months was 56%, and there were 17% treatment failures. The relapse risk was 23% and the mortality 47% [2]. These results were strikingly better when compared to an earlier study where using steroids alone for patients with these disease characteristics resulted in a 5-year survival of only 26% [3]. For many years the combination of cyclosporine and prednisolone was regarded as standard primary treatment of chronic GVHD [4]. However, a more recent large randomized clinical trial comparing the combination therapy with prednisolone monotherapy revealed no difference in the major endpoints therapy-related mortality, overall mortality, need for secondary treatment, or discontinuation of all immunosuppressive therapies. However, disease-free survival was significantly worse in the combination arm, suggesting a destructive  effect of cyclosporine on the graft-versus-leukemia effect [5]. Although this study was performed in patients without thrombocytopenia, it nevertheless did not substantiate the hypothesis that the administration of cyclosporine reduces transplantation-related mortality among patients with chronic GVHD.

Another calcineurin inhibitor used in transplantation is tacrolimus. However, data on its use in chronic GVHD is scarce. Its efficacy as salvage therapy in chronic GVHD was evaluated in a small phase 2 study. Although 13% of patients achieved complete remission of GVHD and could discontinue tacrolimus, a failure rate of 79% is not very encouraging. Nevertheless, it might be of benefit in some selected patients [6].

Cytotoxic drugs (MTX, Cytoxan, Azathioprine, MMF, Pentostatin)

Since low-dose methotrexate (MTX) has been proven to be effective in the prophylaxis of acute GVHD and has low toxicity profile as well as being a cost effective alternative, it has been evaluated in a series of small retrospective analyses for the treatment of chronic GVHD [7-10]. The response rates varied from 59–76%, thus suggesting methotrexate in doses of 7.5 to 15 mg to be active in chronic GVHD. Best responses have been seen in lichenoid, cutaneous, and gastrointestinal chronic GVHD. Consequently, low-dose methotrexate should be evaluated in prospective clinical trials.

Another cytotoxic drug evaluated in a small patient series for the treatment of chronic GVHD is cytoxan. It was used as pulse therapy (1000 mg/m2) and the response was dependent on the organs involved, with best responses seen in skin and mucous membrane GVHD. Remarkably, 80% of patients with liver GVHD also responded to this treatment [11, 12].

The use of azathioprine in addition to prednisolone has been evaluated in a randomized trial in patients with chronic GVHD without thrombocytopenia [3]. No benefit could be observed by the addition of azathioprine. On the contrary, in the combination arm the non-relapse mortality doubled and overall survival decreased from 61% in the prednisolone arm to 47% in the prednisolone plus azathioprine arm. Therefore for treatment of chronic GVHD no role for azathioprine has been as yet established.

The more lymphocyte-selective purine antagonist mycophenolate mofetil (MMF) has been studied in a small prospective and several small retrospective studies for treatment of refractory chronic GVHD [13-15]. The response rates varied from 64-100% depending on organ involvement, with best responses seen in cutaneous manifestations. Generally the drug was well tolerated and a steroid sparing effect was observed in most studies. However, infectious complications seemed to be increased. A recent prospective randomized trial for initial treatment of chronic GVHD has been closed prematurely, as an interim analysis showed no benefit of adding MMF to standard treatment [16]. Therefore MMF should not be added to the initial systemic treatment regimen for chronic GVHD. Nevertheless MMF can be considered for secondary treatment of refractory patients.

Georgia Vogelsang has evaluated the purine analog pentostatin in a prospective phase 2 trial for treatment of steroid-resistant chronic GVHD [17]. Pentostatin was administered at a dosage of 4 mg/m2 every other week for a minimum of 24 weeks. Although most patients had failed more than one prior immunosuppressive regimen (median 4), 55% of the patients had an objective response and survival at 2 years was 70%. Responses were best in lichenoid skin GVHD. Of the 32 responders 8 went off the study because of adverse events. Infections were the most significant toxicity.

Thoraco-abdominal Irradiation

Low dose thoraco-abdominal irradiation (1 Gy) was applied by the group of the Hospital Saint-Louis in Paris to 41 patients with refractory extensive chronic GVHD [18]. Eighty-two percent of the patients achieved a clinical response, and two years after the irradiation 25% of the patients had a complete response. Best responses were seen in patients with fasciitis (79%) and oral GVHD lesions (73%). However, one third of the responders responded only temporarily. The major adverse event was mild pancytopenia. Especially in patients with fasciitis and oral GVHD lesions low dose thoraco-abdominal irradiation may be considered as a safe and efficient treatment option.

Immunomodulatory drugs

Thalidomide was first reported in 1992 to be active in chronic GVHD [19]. In the following years a couple of retrospective studies seemed to confirm its efficacy with response rates between 36 and 75% [20-22]. However, two randomized trials failed to prove any benefit of adding thalidomide to standard treatment of extensive chronic GVHD [23, 24]. The Seattle trial could not even evaluate the efficacy since more than 90% of the patients discontinued the study drug before resolution of GVHD, because of severe neutropenia and neurological symptoms. Therefore the use of thalidomide can not be generally recommended. However, selected patients may benefit from low dose thalidomide (100–200mg/d).

Clofazimine, a drug with activity in leprosy and various chronic autoimmune skin disorders, has been reported to be active also in chronic GVHD in a small phase 2 trial [25]. It was given orally once a day at a dose of 300 mg for 90 days and at a dose of 100 mg thereafter. More than 50% of the patients with skin involvement, contractures or oral GVHD manifestations achieved responses. The most common side effects were gastrointestinal and hyperpigmentation. Therefore clofazimine appears safe and, maybe, efficacious. However, the results of this small study are still to be confirmed in larger trials.

The malaria drug hydroxychloroquine is supposed to interfere with antigen presentation and has synergistic activity with calcineurin inhibitors in vitro. Therefore it has been evaluated for treatment of chronic GVHD in a small phase 2 trial [26]. Forty patients with steroid resistant or dependent chronic GVHD were treated with 12 mg/kg/day with an overall response rate of 53%. In all responders steroids could be tapered to at least 50% of the initial dosage. There were no major toxicities. While this study also awaits confirmation in larger trials, the use of hydroxychloroquine as prophylaxis for chronic GVHD revealed disappointing results in a randomized double blind clinical trial [27].

Cytokine modulation may be another treatment option to treat chronic GVHD. And, indeed, there have been anecdotal reports on the use of soluble tumor-necrosis-factor receptor [28, 29] or of antibodies against the interleukin-2 receptor [30, 31] for treatment of chronic GVHD. With patient numbers between 4 and 10 the response rates varied from 50% to 75%.

Two recent small clinical studies have suggested the tyrosine kinase inhibitor imatinib mesylate to be active in the treatment of steroid-refractory sclerodermatous GVHD, via inhibition of fibroblast activity and TGF-ß [32, 33]. Response rates of 50–79% have been reported. However, especially at higher doses, up to 30% of patients did not tolerate imatinib and stopped treatment. 

Rituximab

As B-cells are involved in the pathophysiology of chronic GVHD via production of allo- and auto-antibodies it seems reasonable that the B-cell specific monoclonal antibody rituximab may be efficacious in the treatment of chronic GVHD. Several studies have addressed the role of rituximab in the treatment of chronic GVHD [34-37]. The overall response rates varied from 50–80%. Organ-specific response rates were 13 to 100% for involvement of the skin, 0 to 83% for involvement of the oral mucosa, 0 to 66% for involvement of the liver, and 0 to 38% for GVHD of the lung.

Extracorporal photopheresis (ECP)

In ECP a small part of the patient’s mononuclear cells are exposed to the photosensitizer 8-methoxypsoralen and irradiated with ultraviolet A light extra-corporally. The irradiated T-cells undergo apoptosis and the apoptotic cells are reinfused into the patient, and it has been suggested that secondarily tolerogenic antigen-presenting cells (APCs) are induced. Therefore ECP therapy, unlike other immunosuppressive regimens, does not cause global immunosuppression, but induces immune tolerance. Recent clinical and animal studies demonstrate that ECP therapy induces antigen-specific regulatory T cells, including CD4+CD25+FoxP3+ T cells and IL-10-producing Tr1 cells [38]. The clinical use of ECP to treat chronic GVHD was pioneered by Hildegard Greinix in Vienna [39]. In 15 patients ECP was well tolerated and achieved complete responses in 80 percent of cutaneous manifestations and 70% for liver involvement. Subsequently ECP was evaluated in a number of clinical trials [40-45]. In general, skin manifestations including sclerodermatous lesions responded best to ECP and occasionally it was also reported to improve pulmonary manifestations of chronic GVHD [46]. In a recent randomized multi-centre trial the addition of ECP to the standard treatment seemed to be more effective even in sclerodermatous skin disease with a 40% response rate at 12 treatment weeks, although the primary endpoint, blinded assessment of skin score, did not reach a significant level [47] However, a clear steroid sparing effect could be demonstrated.

Inhibitors of the mammalian target of rapamycin (mTOR-I)

Sirolimus and everolimus, inhibitors of the mammalian target of rapamycin (mTOR-I), combine immunosuppressive properties with antiproliferative effects on fibroblasts and smooth muscle cells [48]. mTOR-I exert their action by binding to FK-binding protein 12 (FKBP12), and subsequently forming a complex with the mammalian target of rapamycin (mTOR) and the raptor/rictor proteins. The generation of this complex results in cell cycle arrest in G1 via inhibition of DNA transcription, DNA translation, and protein synthesis. In contrast to CNI, sirolimus promotes the generation of CD4+CD25+FoxP3+ regulatory T-cells [49]. These data indicate that mTOR-I could provide additional advantage for the treatment of chronic GvHD both because of their antifibrotic activity and by possibly inducing tolerance. The mTOR-I sirolimus and everolimus have been extensively studied as immunosuppressants in solid organ transplantation. Substituting CNI with mTOR-I seems to overcome long term threats, like chronic allograft dysfunction and vasculopathy after solid organ transplantation [50]. In allogeneic hematopoietic cell transplantation, mTOR-I have demonstrated efficacy in prophylaxis and treatment of acute GVHD in a number of studies [51, 52].

However, considerable toxicity like transplant associated microangiopathy has been observed when mTOR-I were used in combination with CNI, which could be avoided in a CNI-free regimen [53]. Sirolimus has also been evaluated in second line treatment of chronic GVHD in small phase 2 trials mostly in combination with CNI [54-56]. The response rates varied between 56% and 81%. Major adverse events were hyperlipidemia, renal dysfunction, cytopenias and transplant-associated microangiopathy, which lead to termination of therapy in up to 1/3rd of treated patients. These experiences prompted us to avoid the combination of CNI and mTOR-I in the treatment of chronic GVHD. Intriguingly, when analyzing our data on the use of mTOR-I in sclerodermatous chronic GVHD, similar efficacy was achieved despite the absence of calcineurin-inhibitors (CNI). Compared with a study describing the treatment of chronic GVHD with sirolimus in combination with tacrolimus and corticosteroids we achieved similar response rates (76% in our study vs. 73% reported by Couriel et al.) and a more favorable toxicity profile [57]. Importantly, in contrast to the use of CNI, no increased relapse rate has been observed. This suggests that the graft versus leukemia effect is not compromised by mTOR-I therapy but may be even facilitated by the antitumoral activity of mTOR-I [58]. No differences were seen between sirolimus or everolimus treated patients. Importantly, in our study, we observed no nephrotoxicity and TMA was rare (5.9%), correlating with high trough levels of mTOR-I. Generally, if low therapeutic trough levels (4–8 ng/ml) were maintained, toxicities associated with mTOR-I therapy were moderate. Since mTOR-I possibly interfere with wound healing [59], they should be used with caution in patients with cutaneous or mucosal ulcers. In case of progressive ulcerous lesions, other therapeutic modalities should be chosen (e.g. extracorporal photopheresis). mTOR-I seem to enhance plasmatic coagulation, as suggested by significant shortening of prothrombin time in a significant number of patients. Therefore, plasmatic hemostasis markers should be monitored during mTOR-I therapy and antithrombotic prophylaxis should be considered, especially if patients have additional risk factors, e.g. steroid therapy. The involvement of mTOR in coagulation signaling cascades has as yet not been reported. Thus, further experimental studies are needed to clarify the possible role of mTOR-dependent downstream pathways in hemostasis. Hyperlipidemia was frequent, but seldom required therapeutic intervention. Similar results were observed in another CNI-free trial for the treatment of chronic GVHD utilizing everolimus in combination with steroids and in part with azathioprine [60]. Taken together mTOR-I appear to be an effective treatment option for chronic GVHD with a low toxicity profile as long as low therapeutic drug trough levels are maintained and combination treatment with CNI is avoided.

Conclusion

Severe chronic graft versus host disease remains the main factor for late morbidity and mortality in long-term survivors after allogeneic hematopoietic cell transplantation. The only established treatment of chronic graft versus host disease is the use of high dose corticosteroids, which is associated with multiple adverse reactions. Despite a myriad of alternative or additive treatment options up to date no clear strategy to treat chronic GVHD has been established. A better understanding of the pathophysiology of chronic GVHD may guide us in the future to a more sophisticated treatment strategy. Moreover, current available treatment options have to be evaluated in controlled prospective clinical trials. The NIH consensus of diagnosis and staging of chronic GVHD provides the tools for standardized evaluation of different treatment strategies. Most currently available treatment options rely on intensification of immunosuppression at the cost of a higher infection and possibly also a higher relapse rate. However, treatment options focusing on the induction of tolerance have emerged in recent years. While preserving the graft versus leukemia effect, both the use of ECP and the introduction of mTOR-I in the therapy of chronic GVHD have been associated with enhanced formation of regulatory T-cells, thus indicating a tolerance inducing effect.

It seems to be the time to shift the paradigm of treating chronic GVHD from mere immunosuppression to more sophisticated strategies.

References

1. Lee SJ, Klein JP, Barrett AJ, Ringden O, Antin JH, Cahn JY, et al. Severity of chronic graft-versus-host disease: association with treatment-related mortality and relapse. Blood. 2002;100(2):406-414.

2. Sullivan KM, Witherspoon RP, Storb R, Deeg HJ, Dahlberg S, Sanders JE, et al. Alternating-day cyclosporine and prednisone for treatment of high-risk chronic graft-v-host disease. Blood. 1988;72(2):555-561.

3. Sullivan KM, Witherspoon RP, Storb R, Weiden P, Flournoy N, Dahlberg S, et al. Prednisone and azathioprine compared with prednisone and placebo for treatment of chronic graft-v-host disease: prognostic influence of prolonged thrombocytopenia after allogeneic marrow transplantation. Blood. 1988;72(2):546-554.

4. Vogelsang GB. How I treat chronic graft-versus-host disease. Blood. 2001;97(5):1196-1201.

5. Koc S, Leisenring W, Flowers ME, Anasetti C, Deeg HJ, Nash RA, et al. Therapy for chronic graft-versus-host disease: a randomized trial comparing cyclosporine plus prednisone versus prednisone alone. Blood. 2002;100(1):48-51.

6. Carnevale-Schianca F, Martin P, Sullivan K, Flowers M, Gooley T, Anasetti C, Deeg J, Furlong T, McSweeney P, Storb R, Nash RA. Changing from cyclosporine to tacrolimus as salvage therapy for chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2000;6(6):613-20.

7. Giaccone L, Martin P, Carpenter P, Moravec C, Hooper H, Funke VA, et al. Safety and potential efficacy of low-dose methotrexate for treatment of chronic graft-versus-host disease. Bone Marrow Transplant. 2005;36(4):337-341. doi: 10.1038/sj.bmt.1705022.

8. Huang XJ, Jiang Q, Chen H, Xu L, Liu D, Chen Y, et al. Low-dose methotrexate for the treatment of graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2005;36(4):343-348. doi: 10.1038/sj.bmt.1705034.

9. de Lavallade H, Mohty M, Faucher C, Furst S, El Cheikh J, Blaise D. Low-dose methotrexate as salvage therapy for refractory graft-versus-host disease after reduced-intensity conditioning allogeneic stem cell transplantation. Haematologica. 2006;91(10):1438-1440.

10. Inagaki J, Nagatoshi Y, Hatano M, Isomura N, Sakiyama M, Okamura J. Low-dose MTX for the treatment of acute and chronic graft-versus-host disease in children. Bone Marrow Transplant. 2008(6):571-7. doi: 10.1038/sj.bmt.1705922.

11. Mayer J, Krejci M, Pospisil Z, Doubek M, Janikova A, Zackova D, Racil Z, Smardova L, Navratil M, Kamelander J. Successful treatment of steroid-refractory hepatitic variant of liver graft-vs-host disease with pulse cyclophosphamide. Exp Hematol. 2009;37(6):767-73. pmid: 19463776.

12. Mayer J, Krejcí M, Doubek M, Pospísil Z, Brychtová Y, Tomíska M, Rácil Z. Pulse cyclophosphamide for corticosteroid-refractory graft-versus-host disease. Bone Marrow Transplant. 2005;35(7):699-705. doi: 10.1038/sj.bmt.1704829.

13. Takami A, Mochizuki K, Okumura H, Ito S, Suga Y, Yamazaki H, Yamazaki M, Kondo Y, Asakura H, Nakao S. Mycophenolate mofetil is effective and well tolerated in the treatment of refractory acute and chronic graft-versus-host disease. Int J Hematol. 2006;83(1):80-5. pmid: 16443558.

14. Krejci M, Doubek M, Buchler T, Brychtova Y, Vorlicek J, Mayer J. Mycophenolate mofetil for the treatment of acute and chronic steroid-refractory graft-versus-host disease. Ann Hematol. 2005;84(10):681-5. doi: 10.1007/s00277-005-1070-0.

15. Baudard M, Vincent A, Moreau P, Kergueris MF, Harousseau JL, Milpied N. Mycophenolate mofetil for the treatment of acute and chronic GVHD is effective and well tolerated but induces a high risk of infectious complications: a series of 21 BM or PBSC transplant patients. Bone Marrow Transplant. 2002;30(5):287-95. doi: 10.1038/sj.bmt.1703633.

16. Martin PJ, Storer BE, Rowley SD, Flowers ME, Lee SJ, Carpenter PA, Wingard JR, Shaughnessy PJ, DeVetten MP, Jagasia M, Fay JW, van Besien K, Gupta V, Kitko C, Johnston LJ, Maziarz RT, Arora M, Jacobson PA, Weisdorf D. Evaluation of mycophenolate mofetil for initial treatment of chronic graft-versus-host disease. Blood. 2009 May 21;113(21):5074-82. doi: 10.1182/blood-2009-02-202937.

17. Jacobsohn DA, Chen AR, Zahurak M, Piantadosi S, Anders V, Bolanos-Meade J, et al. Phase II study of pentostatin in patients with corticosteroid-refractory chronic graft-versus-host disease. J Clin Oncol. 2007;25(27):4255-4261. doi: 10.1200/JCO.2007.10.8456.

18. Robin M, Guardiola P, Girinsky T, Hernandez G, Esperou H, Ribaud P, et al. Low-dose thoracoabdominal irradiation for the treatment of refractory chronic graft-versus-host disease. Transplantation. 2005;80(5):634-642. pmid: 16177638.

19. Vogelsang GB, Farmer ER, Hess AD, Altamonte V, Beschorner WE, Jabs DA, et al. Thalidomide for the treatment of chronic graft-versus-host disease. N Engl J Med. 1992;326(16):1055-1058. pmid: 1549151.

20. Browne PV, Weisdorf DJ, Defor T, Miller WJ, Davies SM, Filipovich A, et al. Response to thalidomide therapy in refractory chronic graft-versus-host disease. Bone Marrow Transplant. 2000;26(8):865-869.

21. Kulkarni S, Powles R, Sirohi B, Treleaven J, Saso R, Horton C, Atra A, Ortin M, Rudin C, Goyal S, Sankpal S, Meller S, Pinkerton CR, Mehta J, Singhal S. Thalidomide after allogeneic haematopoietic stem cell transplantation: activity in chronic but not in acute graft-versus-host disease. Bone Marrow Transplant. 2003;32(2):165-70.

22. van de Poel MH, Pasman PC, Schouten HC. The use of thalidomide in chronic refractory graft versus host disease. Neth J Med. 2001;59(2):45-9. doi: 10.1016/S0300-2977(01)00133-4.

23. Arora M, Wagner JE, Davies SM, Blazar BR, Defor T, Enright H, et al. Randomized clinical trial of thalidomide, cyclosporine, and prednisone versus cyclosporine and prednisone as initial therapy for chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2001;7(5):265-273.

24. Koc S, Leisenring W, Flowers ME, Anasetti C, Deeg HJ, Nash RA, et al. Thalidomide for treatment of patients with chronic graft-versus-host disease. Blood. 2000;96(12):3995-3996.

25. Lee SJ, Wegner SA, McGarigle CJ, Bierer BE, Antin JH. Treatment of chronic graft-versus-host disease with clofazimine. Blood. 1997;89(7):2298-2302.

26. Gilman AL, Chan KW, Mogul A, Morris C, Goldman FD, Boyer M, et al. Hydroxychloroquine for the treatment of chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2000;6(3A):327-334. pmid: 10905770.

27. Fong T, Trinkaus K, Adkins D, Vij R, Devine SM, Tomasson M, Goodnough LT, Lopez S, Graubert T, Shenoy S, Dipersio JF, Khoury HJ. A randomized double-blind trial of hydroxychloroquine for the prevention of chronic graft-versus-host disease after allogeneic peripheral blood stem cell transplantation. Biol Blood Marrow Transplant. 2007;13(10):1201-6.

28. Busca A, Locatelli F, Marmont F, Ceretto C, Falda M. Recombinant human soluble tumor necrosis factor receptor fusion protein as treatment for steroid refractory graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. Am J Hematol. 2007;82(1):45-52.

29. Chiang KY, Abhyankar S, Bridges K, Godder K, Henslee-Downey JP. Recombinant human tumor necrosis factor receptor fusion protein as complementary treatment for chronic graft-versus-host disease. Transplantation. 2002 Feb 27;73(4):665-7. pmid: 11889452.

30. Teachey DT, Bickert B, Bunin N. Daclizumab for children with corticosteroid refractory graft-versus-host disease. Bone Marrow Transplant. 2006;37(1):95-9. doi: 10.1038/sj.bmt.1705199.

31. Willenbacher W, Basara N, Blau IW, Fauser AA, Kiehl MG. Treatment of steroid refractory acute and chronic graft-versus-host disease with daclizumab. Br J Haematol. 2001;112(3):820-3. doi: 10.1046/j.1365-2141.2001.02582.x.

32. Magro L, Mohty M, Catteau B Magro L, Catteau B, Coiteux V, Bruno B, Jouet JP, Yakoub-Agha I. Imatinib mesylate as salvage therapy for refractory sclerotic chronic graft-versus-host disease. Bone Marrow Transplant. 2008 Dec;42(11):757-60.

33. Olivieri A, Locatelli F, Zecca M, Sanna A, Cimminiello M, Raimondi R, Gini G, Mordini N, Balduzzi A, Leoni P, Gabrielli A, Bacigalupo A. Imatinib for refractory chronic graft-versus-host-disease with fibrotic features. Blood. 2009 Jul 16;114(3):709-18. doi: 10.1182/blood-2009-02-204156.

34. Cutler C, Miklos D, Kim HT, Treister N, Woo SB, Bienfang D, et al. Rituximab for steroid-refractory chronic graft-versus-host disease. Blood. 2006;108(2):756-762. doi: 10.1182/blood-2006-01-0233.

35. Ratanatharathorn V, Ayash L, Reynolds C, Silver S, Reddy P, Becker M, et al. Treatment of chronic graft-versus-host disease with anti-CD20 chimeric monoclonal antibody. Biol Blood Marrow Transplant. 2003;9(8):505-511.

36. Zaja F, Bacigalupo A, Patriarca F, Stanzani M, Van Lint MT, Filì C, Scimè R, Milone G, Falda M, Vener C, Laszlo D, Alessandrino PE, Narni F, Sica S, Olivieri A, Sperotto A, Bosi A, Bonifazi F, Fanin R; GITMO (Gruppo Italiano Trapianto Midollo Osseo). Treatment of refractory chronic GVHD with rituximab: a GITMO study. Bone Marrow Transplant. 2007;40(3):273-7. doi: 10.1038/sj.bmt.1705725.

37. von Bonin M, Oelschlägel U, Radke J, Stewart M, Ehninger G, Bornhauser M, Platzbecker U. Treatment of chronic steroid-refractory graft-versus-host disease with low-dose rituximab. Transplantation. 2008;86(6):875-9. pmid: 18813113.

38. Xia CQ, Campbell KA, Clare-Salzler MJ. Extracorporeal photopheresis-induced immune tolerance: a focus on modulation of antigen-presenting cells and induction of regulatory T cells by apoptotic cells.Curr Opin Organ Transplant. 2009;14(4):338-43.

39. Greinix HT, Volc-Platzer B, Rabitsch W, Gmeinhart B, Guevara-Pineda C, Kalhs P, Krutmann J, Hönigsmann H, Ciovica M, Knobler RM. Successful use of extracorporeal photochemotherapy in the treatment of severe acute and chronic graft-versus-host disease. Blood. 1998;92(9):3098-104.

40. Seaton ED, Szydlo RM, Kanfer E, Apperley JF, Russell-Jones R. Influence of extracorporeal photopheresis on clinical and laboratory parameters in chronic graft-versus-host disease and analysis of predictors of response. Blood. 2003;102(4):1217-23.

41. Bisaccia E, Palangio M, Gonzalez J, Adler KR, Rowley SD, Goldberg SL. Treating refractory chronic graft-versus-host disease with extracorporeal photochemotherapy. Bone Marrow Transplant. 2003;31(4):291-4. doi: 10.1038/sj.bmt.1703830.

42. Ilhan O, Arat M, Arslan O, Ayyildiz E, Sanli H, Beksac M, Ozcan M, Gürman G, Akan H. Extracorporeal photoimmunotherapy for the treatment of steroid refractory progressive chronic graft-versus-host disease. Transfus Apher Sci. 2004;30(3):185-7. doi: 10.1016/j.transci.2004.02.003.

43. Foss FM, DiVenuti GM, Chin K, Sprague K, Grodman H, Klein A, Chan G, Stiffler K, Miller KB. Prospective study of extracorporeal photopheresis in steroid-refractory or steroid-resistant extensive chronic graft-versus-host disease: analysis of response and survival incorporating prognostic factors. Bone Marrow Transplant. 2005;35(12):1187-93. doi: 10.1038/sj.bmt.1704984.

44. Rubegni P, Cuccia A, Sbano P, Cevenini G, Carcagnì MR, D'Ascenzo G, De Aloe G, Guidi S, Guglielmelli P, Marotta G, Lauria F, Bosi A, Fimiani M. Role of extracorporeal photochemotherapy in patients with refractory chronic graft-versus-host disease. Br J Haematol. 2005;130(2):271-5.

45. Couriel DR, Hosing C, Saliba R, Shpall EJ, Anderlini P, Rhodes B, Smith V, Khouri I, Giralt S, de Lima M, Hsu Y, Ghosh S, Neumann J, Andersson B, Qazilbash M, Hymes S, Kim S, Champlin R, Donato M. Extracorporeal photochemotherapy for the treatment of steroid-resistant chronic GVHD. Blood. 2006;107(8):3074-80. doi: 10.1182/blood-2005-09-3907.

46. Child FJ, Ratnavel R, Watkins P, Samson D, Apperley J, Ball J, Taylor P, Russell-Jones R. Extracorporeal photopheresis (ECP) in the treatment of chronic graft-versus-host disease (GVHD). Bone Marrow Transplant. 1999;23(9):881-7.

47. Flowers ME, Apperley JF, van Besien K, Elmaagacli A, Grigg A, Reddy V, Bacigalupo A, Kolb HJ, Bouzas L, Michallet M, Prince HM, Knobler R, Parenti D, Gallo J, Greinix HT. A multicenter prospective phase 2 randomized study of extracorporeal photopheresis for treatment of chronic graft-versus-host disease. Blood. 2008;112(7):2667-74. doi: 10.1182/blood-2008-03-141481.

48. Kirken RA, Wang YL. Molecular actions of sirolimus: sirolimus and mTor. Transplant. Proc. 2003;35:227S-230S. pmid: 12742500.

49. Zeiser R, Nguyen VH, Beilhack A, Buess M, Schulz S, Baker J, et al. Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production. Blood. 2006;108:390-399. doi: 10.1182/blood-2006-01-0329.

50. Chapman JR, Valantine H, Albanell J, Arns WA, Campistol JM, Eisen H, Frigerio M, Lehmkuhl H, Marcen R, Morris R, Nashan B, Pascual J, Pohanka E, Segovia J, Zuckermann A. Proliferation signal inhibitors in transplantation: questions at the cutting edge of everolimus therapy. Transplant Proc. 2007;39(10):2937-50. doi: 10.1016/j.transproceed.2007.09.008.

51. Cutler C, Li S, Ho VT, Koreth J, Alyea E, Soiffer RJ, et al. Extended follow-up of methotrexate-free immunosuppression using sirolimus and tacrolimus in related and unrelated donor peripheral blood stem cell transplantation. Blood. 2007;109:3108-3114. doi: 10.1182/blood-2006-09-046219.

52. Benito AI, Furlong T, Martin PJ, Anasetti C, Appelbaum FR, Doney K, et al. Sirolimus (rapamycin) for the treatment of steroid-refractory acute graft-versus-host disease. Transplantation. 2001;72:1924-1929. pmid:  11773890.

53. Schleuning M, Judith D, Jedlickova Z, Stübig T, Heshmat M, Baurmann H, Schwerdtfeger R. Calcineurin inhibitor-free GVHD prophylaxis with sirolimus, mycophenolate mofetil and ATG in allo-SCT for leukemia patients with high relapse risk: an observational cohort study. Bone Marrow Transplant. 2009;43:717-723. doi: 10.1038/bmt.2008.377.

54. Johnston LJ, Brown J, Shizuru JA, Stockerl-Goldstein KE, Stuart MJ, Blume KG, et al. Rapamycin (sirolimus) for treatment of chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2005;11:47-55.

55. Couriel DR, Saliba R, Escalon MP, Hsu Y, Ghosh S, Ippoliti C, et al. Sirolimus in combination with tacrolimus and corticosteroids for the treatment of resistant chronic graft-versus-host disease. Br J Haematol. 2005;130:409-417. doi: 10.1111/j.1365-2141.2005.05616.x.

56. Jurado M, Vallejo C, Pérez-Simón JA, Brunet S, Ferra C, Balsalobre P, et al. Sirolimus as part of immunosuppressive therapy for refractory chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2007;13:701-706.

57. Jedlickova Z, Burlakova I, Cook A, Baurmann H, Schwerdtfeger R, Schleuning M. mTOR inhibitors for treatment of sclerodermatous chronic graft-versus-host disease following allogeneic stem cell transplantation. Bone Marrow Transplant. 2009;43(suppl. 1):S123(abs.).

58. Recher C, Beyne-Rauzy O, Demur C, et al. Antileukemic activity of rapamycin in acute myeloid leukemia. Blood. 2005;105:2527-2534. doi: 10.1182/blood-2004-06-2494.

59. Kuppahally S, Al-Khaldi A, Weisshaar D, et al. Wound healing complications with de novo sirolimus versus mycophenolate mofetil-based regimen in cardiac transplant recipients. Am.J.Transplant. 2006;6:986-992.

60. Klink A, Schilling K, Rapp K, Höffken K, Sayer HG. High overall response rate in calcineurin inhibitor-free treatment with the mTOR inhibitor everolimus in advanced extensive chronic GvHD after allogeneic stem cell transplantation. Blood. 2008;112:2210 (abs.).

Volume 2, Number 2(6)
03/01/2011

Download PDF version

doi 10.3205/ctt-2009-en-000050.01
Submitted 23 August 2009
Accepted 08 October 2009
Published 16 October 2009

Back to the list