Role of Hematopoietic Stem Cell Transplantation in Relapsed/Refractory Hodgkin Lymphoma


Angelo Michele Carella

Direttore U.O. Ematologia, IRCCS San Martino, Genova


Correspondence to: Pr. Angelo Michele Carella. E-mail: angelomichele.carella@hsanmartino.it

Published: October 2, 2012
Received: July 19, 2012
Accepted: September 21, 2012
Meditter J Hematol Infect Dis 2012, 4(1): e2012059, DOI 10.4084/MJHID.2012.059
This article is available on PDF format at:
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited


Abstract

Hodgkin lymphoma is one of the most curable human tumors. Despite this, about 30% of these patients relapsed or are primary refractory to the first line treatment. Autografting is generally considered the standard of care for these patients. Alternative salvage strategies have been evaluated such as high dose sequential and tandem autografting strategies. In younger patients, refractory or early relapsed after autografting, allogeneic stem cell transplantation has been employed but this approach has been followed by significant concerns since the treatment related mortality, often exceeded 40-50%, and relapses were not uncommon. It is clear that patient selection remains an issue in all allografting reports.
At the end, new drugs and novel treatment strategies, that are based on our understanding of the disease biology and signaling pathways, are needed to improve treatment outcome for these patients. The two leading compounds Brentuximab Vedotin and Panobinostat, are currently under evaluation in several clinical trials.


Introduction
Treatment outcome for patients with Hodgkin lymphoma has steadly improved over the last half-century. Today, about 70-80% of patients can be cured with modern treatment strategy. Unfortunately, some patients fail to attain an initial remission and approximately 30% of those, who achieve remission, relapse. Salvage chemotherapy followed by high-dose chemotherapy (HDC) and autologous stem cell transplantation (ASCT) has become the first choice in relapsed/refractory patients. Alternative strategies such as allografting, new cytostatic drugs and biological agents with proven efficacy in preclinical model may improve the outcome of relapsed/refractory patients to ASCT.

Salvage Chemotherapy Treatments Before ASCT
The ideal salvage regimen should produce: a) high response rate, b) acceptable hematologic and nonhematologic toxicity and c) not impair the ability to mobilize peripheral blood stem cell mobilization. There is no consensus on the gold-standard of salvage chemotherapy. Regimens such as IGEV, DHAP, ICE or GDP are reasonable options but no direct comparison among them is available. In our opinion, patients who are non responders to platinum and/or gemcitabin containing regimens and have stable disease, can safely proceed to ASCT without any kind of further previous chemotherapy. An alternative salvage regimen could be used in patients with progressive disease and/or larger volume disease but the results are generally worse. It has recommended to use a salvage chemotherapy with which clinicians are confortable that results in acceptable toxicity, high response rates and good stem cell mobilization.

Autologous Stem Cell Transplantation
At the end of ’80th, the Italian teams pioneered ASCT in 50 patients with advanced Hodgkin lymphoma.[1] Soon after, a number of international Phase II clinical trials have confirmed the feasibility and the efficacy of this procedure alone. In all these retrospective studies, the results with ASCT appeared superior to those achieved with conventional salvage therapy alone. Unfortunately, the studies had a relatively small sample size and needed to be tested in controlled clinical trials to better clarify the role of ASCT. The first randomized trial comparing HDC/ASCT versus standard-dose salvage chemotherapy was carried out within the British National Lymphoma Investigation.[2] As known, the study was stopped earlier because of an event-free survival advantage for the HDC arm (53% versus 10%; p=0.025). This study showed that HDC produced a better response rate, but, it did not answer the crucial question if was better to utilize ASCT in early or delayed phases of disease. The second controlled study was organized by the German Hodgkin Study Group (GHSG) in cooperation with our Unit in Genoa.[3] They carried out a randomized trial in chemotherapy-sensitive patients comparing HDC (BEAM) vs conventional chemotherapy (two courses of Dexa-BEAM). At a median follow-up of 39 months, freedom from treatment failure was significantly better for the patients in the transplant arm (55% vs 34%, p=0.019) and, most importantly, the significant benefit from ASCT was confirmed in early and late first relapse. In UK and German trials, the overall survival did not differ in the two treatments. These results have been confirmed in other two case-control studies.[4,5] The conclusion of all these trials was that HDC/ASCT is a superior treatment for patients with relapsed or refractory Hodgkin lymphoma and this approach has become the standard therapy for these patients.
In recent years, the ASCT techniques have been greatly improved, prognostic factors have been identified, and the optimal timing of ASCT has been evaluated. A new opportunity to further improve the ASCT procedure was to intensify the therapy with the use of augmented doses of mobilization regimens[6] or additional therapy after PBSC collection.[7] Both these techniques have been reported to be able to improve the outcome. The Cologne high-dose sequential (HDS) protocol utilized 2 cycles of conventional chemotherapy. The patients, who achieved a response, received high-dose cyclophosphamide with subsequent PBSC collection; soon after, high-dose methotrexate and etoposide and, finally, HDC (BEAM) and ASCT was given to the patients. The authors were able to demonstrate that HDS was feasible with acceptable toxicity.[8] Recently, the German Hodgkin Study Group and EBMT reported the HD-R2 trial, a randomized comparison of high-dose sequential therapy followed by ASCT vs standard chemotherapy (DHAP) followed by ASCT. The median follow-up was of 42 months and no significant difference in freedom from therapy failure, progression free survival or overall survival was observed until now.[9] In both randomized studies, the results in primary refractory patients were disappointing. Sweetenham et al. published a retrospective EBMT analysis in 175 patients who fail to enter remission with induction chemotherapy.[10] The actuarial 5-year PFS and overall survival was 32% and 36%, respectively; moreover, patients receiving more than one chemotherapy line before ASCT did worse in terms of overall survival and progression-free survival. Similar results were achieved by GELTAMO Cooperative Group and Fermč in France in 62 and 157 patients, respectively.[11,12] The results were better in the ABMTR analysis on 122 patients not responsive to first line therapy.[13] The reasons of these discrepancies are not clear. Unfortunately, patients with rapidly progressive disease, poor performance status, older age, and poor stem cell harvest were not included in these reports; moreover, it is possible that these different results were subject to significant selection of patients.

Can We Improve the Results after Autografting?
An intensification strategy has been tested with the use of double ASCT.[14] There are limited registry data to support such a strategy. In the GELA multicenter H96 trial the patients were assigned to a single or double ASCT on the basis of presence of risk factors. The patients with primary refractory disease or with at least 2 poor risk factors, received double ASCT; all others received a single ASCT.[15] The overall survival at 5 years was 46% in the poor risk patients with 6% transplant-related mortality. The IBMTR reported 21 Hodgkin lymphoma patients underwent a second ASCT.[16] No difference was found in outcome; the results were particularly worse in patients receiving the second ASCT within 12 months from the first ASCT. Transplant-related mortality was 11% with the double ASCT and 5-year PFS was 30%. These trials demonstrated to be feasible but should be tested prospectively and compared with a standard single ASCT.

Prognosis after Failure ASCT
The median overall survival for patients who progress or relapse after ASCT is 2 years or less. The GELTAMO cooperative group reported the long-term outcome in 175 patients who relapsed at a median time of 10 (range, 4-125) months after ASCT. One third and one fourth of them are alive and free of relapse at 3 years.[17] Adverse prognostic factors for progression-free survival were advanced stage at relapse and short time interval between ASCT and relapse less than 1 year. The patients who had both characteristics had 14% 3-year progression-free survival. Another analysis derived by Lymphoma Working Party of the EBMT database on 462 patients. After a median follow-up of 49 months, overall survival was 32% at 5-years. In multivariate analysis independent risk factors for overall survival were: advanced stage, poor performance status, age >50 years, early relapse. Overall survival was extremely low (12%) in patients with 2 or more risk factors while was high in 62% of patients with one factor only.[18]

The Role of Functional Imaging Before ASCT
Retrospective studies suggest that Gallium or FDG-PET after salvage chemotherapy and before HDC/ASCT can be predictive of poor outcome.[19] More recently, Moskowitz et al. have studied functional imaging after salvage chemotherapy reporting 75% 5-years event-free survival if functional imaging was negative and 31% for functional imaging-positive disease.[20] Castagna et al. in 24 patients who underwent FDG-PET after 2 cycles of salvage chemotherapy, reported 2-years PFS of 93% for PET-negative and 10% for PET-positive patients.[21] More recently, the role of PET has been analyzed in a group of 101 patients who received ASCT for Hodgkin lymphoma and non-Hodgkin lymphomas. Both FDG-PET after 2 courses of chemotherapy and clinical risk score were independent prognostic factors for progression-free survival after ASCT;[22] moreover a poor outcome after ASCT was identified by the Houston team when PET resulted positive before ASCT.[23] Despite these and other reports, PET scanning is still contradictory to predict outcome after ASCT. We need more robust results in order to determinate risk-adapted therapy outside of a clinical trial.

Novel Agents
After more than 30 years of absence of new drugs in this disease, in these last years several compounds have been identify as promising agents, mainly for the treatment of patients with relapsed HL. The two leading compounds, Brentuximab Vedotin and Panobinostat, are currently employed with excellent results. Brentuximab Vedotin has able to achieve 75% of overall response rate (34% CR) with a median duration of response for remitters patients of 25%.[24] The potent pan- deacytilase inhibitor (Panobinostat) was able to determine an overall objective response of 27% of relapsed/refractory HL patients.[25] Although the patients enrolled onto these studies had a poor prognosis and were previously highly pretreated with multiple chemotherapy regimens, almost all patients achieved tumor regression, mainly with Brentuximab Vedotin. A new strategy to improve autografting might be to incorporate these drugs with salvage regimen and/or as maintenance after stem cell transplantation. The new randomized trials with these drugs are ongoing and hopefully can modify the number of relapses. In figure 1 it has been showed the possible future perspectives employments of these drugs.


Figure 1.
How to integrate targeted therapies into clinical practice for the treatment of Hodgkin Lymphoma.


Allogeneic Stem Cell Transplantation (AlloSCT)
Registry-based studies published in 1996 with myeloablative allografting gave disappointing results. AlloSCT was able to result in lower relapse rates but TRM exceeded 50% due to toxicity and infection complications. The explanation for this high mortality is uncertain but might include the selection of very high risk patients (many patients were allografted in advanced phase of the disease) combined with immunodeficiency, peculiar to Hodgkin lymphoma, leading to infectious complications. The use of low-dose preparative regimens, aimed at immunosuppression rather that tumor ablation, has decreased the early mortality rate in hematological neoplasias.[26,27] This new approach have become increasingly popular after it became apparent that much of the anticancer effect from allografting is due to the adoptive immunotherapy. The largest cohort of patients (n=285) treated with RIC was recently reported by the Lymphoma Working Party of the EBMT.[28] Patients have been highly pretreated with a median of 4 lines of therapy. The 100-day TRM was 12% but increased to 22% at 3 years. The best results were achieved in patients with chemosensitive disease. The development of chronic GVHD was associated with a higher TRM but a lower relapse rate. In a landmark analysis the development of GVHD by 9 months post-RIC was associated with lower relapse rate. Similar results were recently updated by MD Anderson Cancer Center in 58 patients with relapsed or refractory Hodgkin lymphoma.[19] More recently, the final results of a multicenter phase II prospective study on the role of RIC were presented at ASH2010.[29] Ninety-two patients with an HLA identical sibling or a MUD were treated with two courses of salvage chemotherapy. Seventy-eight patients (85%), who showed no progression, were eligible to receive a RIC. All patients with refractory disease died of lymphoma. Chronic GVHD was associated with a lower relapse incidence and a better progression-free survival. Patients allografted in remission had a significantly better outcome.
Sureda et al. has performed under Lymphoma Working Party of the EBMT the only analysis which to compare outcomes after RIC or myeloablative conditioning.[30] Ninety-seven patients were allografte after RIC and 93 patients after myeloablative regimens. Non-relapse mortality was significantly decreased in the RIC group. Chronic GVHD significantly decreased the incidence of relapse, which translated into better progression-free survival and overall survival. The conclusion of this analysis was that  the significative reduction of the transplant-related mortality after RIC was able to put in evidence the existence of a GVHL, which was able to improve the long-term outcome of relapsed and refractory Hodgkin lymphoma patients. The most direct evidence of a GVHL comes from Hodgkin lymphoma response to donor lymphocyte infusion. Peggs and associates described the results of RIC and donor lymphocyte infusion in patients with multiply relapses. The conditioning regimen included fludarabine, melphalan and alemtuzumab.[31] Patients who had less than a CR or progression at 3 months received donor lymphocyte infusion. All patients engrafted and the 4-year progression free survival rate was 39% with a nonrelapsed mortality at 2 years of 16%. Sixteen patients received donor lymphocyte infusion at 3 months because of lack of response or progression and 8 of them achieved complete remission.
Other small series in Hodgkin lymphoma confirmed these results and reported response rate of 44-54% following DLI administration.[28,31-34] All these data confirm the presence of a clinically effective GVHL effect.

Tandem ASCT and RIC
Autografting and myeloablative allografting each offers potential roles in the treatment of relapsed/refractory Hodgkin lymphoma. Each modality has its limitations: ASCT has become the standard therapy for these patients but the relapses remain the most important cause of treatment failure; allografting is conditioned by high-mortality risks mediated by immunosuppressive drugs and GVHD. The safety of allografting has improved with the use of RIC, but, since graft vs Hodgkin lymphoma responses might be insufficient when Hodgkin lymphoma is bulky and tumor growth is rapid, it was thought that intensive lymphoma cytoreduction prior to RIC may allow reaction to be exploited.[34] This tactic could provide the benefit of a conventional allograft but with a reduction in the typical acute toxicities and associate mortality of myeloablative therapy. Tandem ASCT/RIC was pioneered by the Genoa team in 10 patients with Hodgkin lymphoma and 5 patients with non-Hodgkin lymphoma.[34] Thirteen of the 15 patients were considered to have poor prognoses due to chemotherapy-refractory disease (n=6), relapse (n=8), or bulky disease at ASCT. The allogeneic conditioning consisted of fludarabine and cyclophosphamide with short-course of methotrexate and cyclosporine post-grafting for GVHD prophylaxis. Nine of 12 patients in partial remission after ASCT achieved complete remission after RICT, while 3 patients had progressive disease. Donor lymphocyte infusions were given to 7 patients, in 2 patients for progressive or persistent disease and in 5 patients for mixed chimerism and persistent disease. At the time of the analysis, 10 patients were alive, while 5 patients died either from progressive disease (n=2), progressive disease and GVHD (n=1), chronic GVHD (n=1) or Aspergillus infection (n=1). The severity of GVHD appeared tolerable with only 1 patient dying directly from complications of extensive chronic GVHD. Recently, we have updated our results together with those of Humanitas Institute of Milan.[35] A total of 27 patients underwent tandem ASCT/RIC. Twelve patients were chemosensitive and 15 chemorefractory at ASCT. The time interval between ASCT and RIC was 3 months. Chimerism studies indicated 100% donor-derived engraftment. Seven patients developed acute GVHD and 9 chronic GVHD (2 limited and 7 extensive). At the last follow up 17 patients (63%) were alive, 12 (70.6%) in CR and 5 (29.6%) with persistent disease. Ten patients expired (37%): 7 patients of disease progression, 1 patient of acute GVHD, 1 patient of extensive chronic GVHD and 1 patient of infection. Overall survival was 47 months at a median follow up of 46 months. These results suggest that GvLy may have a role on residual disease after ASCT. These results have been recently confirmed also by Gutterman et al. in 23 Hodgkin lymphoma patients. The U.K. Authors has employed a more complex approach with a combination of an intensive preparative regimen (BEAM) together with a profound T-cell depletion with Alemtuzumab as acute GVHD prophylaxis. They demonstrated that this approach was associated with sustained donor engraftment, high response rate, minimal TRD (7.6%), and a low incidence of extensive GVHD[35]. In conclusion, the tandem auto-allotransplant is a feasible approach in high-risk Hodgkin lymphoma and in the next future it should be evaluated on more patients, possibly in a randomized size.

Conclusions
Stem cell transplantation is an effective approach in relapsed/refractory patients with Hodgkin lymphoma. Today the patients receive cell collected from peripheral blood in combination with growth factors and a rapid return of normal blood cells is achieved. The procedure is generally safe and, in some instances, can be carried out as an outpatient basis. A new strategy to improve autografting might be to incorporate the new drugs Panobinostat and Brentuximab Vedotin combined with salvage regimens and/or as maintenance after autografting.
The place of allografting in Hodgkin lymphoma remains a point for debate. Allografting has yielded lower relapse rates compared to ASCT, likely due to graft-versus-lymphoma effect. Long-term results of RIC demonstrate a progression free survival of 25-35% and an overall survival at 2-3 years of 35-60%. However, the long-term effects of acute and chronic graft-versus-host disease including both mortality and potentially serious morbidity represent a reason for caution in recommending this treatment approach. On this basis, we recommend RIC in the context of prospective clinical trials. In the next future we should try to reduce the relapse rates after RIC and prospectively compare RIC to both autografting preceeded and followed by Brentuximab Vedotin / Panobinostat.

References

  1. Carella AM, Congiu AM, Gaozza E, Mazza P, Ricci P, Visani G, et al. High-dose chemotherapy with autologous bone marrow transplantation in 50 advanced resistant Hodgkin’s disease patients: an Italian study group report. J Clin Oncol 1988;6:1411-1416 PMid:2458439 
  2. Linch DC, Winfield D, Goldstone AH, Moir D, Hancock B, McMillan A, et al. Dose intensification with autologous bonemarrow transplantation in relapsed and re¬sistant Hodgkin’s disease: results of a BNLI randomized trial. Lancet. 1993;341:1051-4. http://dx.doi.org/10.1016/0140-6736(93)92411-L 
  3. Schmitz N, Pfistner B, Sextro M, Sieber M, Carella AM, Haenel M, et al. Aggressive conventional chemotherapy compared with high-dose chemotherapy with autologous haemopoietic stem-cell transplantation for relapsed chemosensitive Hodgkin’s disease: a randomized trial. Lancet 2002;359:2065-2070. http://dx.doi.org/10.1016/S0140-6736(02)08938-9 
  4. Yuen AR, Rosenber SA, Hoppe RT, Halpern JD, Horning SJ. Comparison between conventional salvage therapy and high-dose therapy with autografting for recurrent or refractory Hodgkin’s disease. Blood 1997;89:814-822. PMid:9028312 
  5. Andre M, Henry-Amar M, Pico JL, Brice P, Blaise D, Kuentz M, et al. Comparison of high-dose therapy and autologous stem-cell transplantation with conventional therapy for Hodgkin’s disease induction failure: a case-control study. Societe Francaise de Greffe de Moelle. J Clin Oncol 1999;17:222-229. PMid:10458237 
  6. Stewart DA, Guo D, Glück S, Morris D, Chaudhry A, deMetz C, et al. Double high-dose therapy for Hodgkin’s disease with dose intensive cyclophosphamide, etoposide, and cisplatin (DICEP) prior to high-dose melphalan and autologous stem cell transplantation. Bone Marrow Transplant 2000;26:383-388. http://dx.doi.org/10.1038/sj.bmt.1702541  PMid:10982284 
  7. Josting A, Müller H, Borchmann P, Baars JW, Metzner B, Döhner H, et al. Dose intensity of chemotherapy in patients with relapsed Hodgkin’s lymphoma. J Clin Oncol. 2010;28:5074-80. http://dx.doi.org/10.1200/JCO.2010.30.5771  PMid:20975066 
  8. Josting A, Sieniawski M, Glossmann JP, Staak O, Nogova L, Peters N, et al. High-dose sequential chemotherapy followed by autologous stem cell transplantation in relapsed and refractory aggressive non-Hodgkin’s lymphoma: results of a multicenter phase II study. Ann Oncol. 2005;16:1359-65. http://dx.doi.org/10.1093/annonc/mdi248  PMid:15939712 
  9. Josting A. Autologous transplantation in relapsed and refractory Hodgkin’s disease. Eur J Haematol Suppl. 2005;66:141-145. http://dx.doi.org/10.1111/j.1600-0609.2005.00468.x 
  10. Sweetenham JW, Carella AM, Taghipour G, Cunningham D, Marcus R, Della Volpe A, et al. High dose therapy and autologous stem cell transplantation for adult patients with Hodgkin’s disease who fail to enter remission after induction chemotherapy: Results in 175 patients reported to the EBMT. J Clin Oncol. 1999;17:3101-3109. PMid:10506605 
  11. Constans M, Sureda A, Terol M J, Arranz R, Caballero MD, Iriondo A, Jarque I, et al. Autologous stem cell transplantation for primary refractory Hodgkin’s disease: results and clinical variables affecting outcome. Ann Oncol. 2003;14:745-51. http://dx.doi.org/10.1093/annonc/mdg206  PMid:12702529 
  12. Fermé C, Mounier N, Diviné M, Brice P, Stamatoullas A, Reman O, et al. Intensive salvage therapy with high-dose chemotherapy for patients with advanced Hodgkin’s disease in relapse or failure after initial chemotherapy: Results of the Groupe d’Études des Lymphomes de l’adulte H89 trial. J Clin Oncol. 2002;20:467-75. PMid:11786576 
  13. Lazarus HM, Rowlings PA, Zhang M-J, Vose JM, Armitage JO, Bierman PJ, et al. Autotransplants for Hodgkin’s disease in patients never achieving remission: A report from the Autologous Blood and Marrow Transplant Registry. J Clin Oncol. 1999;17:534-45. PMid:10080597 
  14. Fung HC, Stiff P, Schriber J, Toor A, Smith E, Rodriguez T, et al. Tandem autologous stem cell transplantation for patients with primary refractory or poor risk recurrent Hodgkin lymphoma. Biol Blood Marrow Transplantation 2007;13:594-600. http://dx.doi.org/10.1016/j.bbmt.2007.01.072  PMid:17448919 
  15. Morschhauser F, Brice P, Fermč C, Diviné M, Salles G, Bouabdallah R, et al. Risk-adapted salvage treatment with single or tandem autologous stem-cell transplantation for first relapse/refractory Hodgkin’s lymphoma: results of the prospective multicenter H96 trial by the GELA/SFGM Study Group. J Clin Oncol 2008;26:5980-5987. http://dx.doi.org/10.1200/JCO.2007.15.5887  PMid:19018090 
  16. Smith SM, van Besien K, Carreras J, Bashey A, Cairo MS, Freytes CO, et al. Second autologous stem cell transplantation for relapsed lymphoma after a prior autologous transplant. Biol Blood Marrow Transplant 2008;14:904-912. http://dx.doi.org/10.1016/j.bbmt.2008.05.021  PMid:18640574 PMCid:3353768 
  17. Constans M, Sureda A, Arranz R, Caballero MD, Carreras E, Conde E, et al. Relapse after autologous stem cell transplantation for Hodgkin’s lymphoma: prognostic factors affecting long-term outcome. Eur J Haematol. 2004;73:53. 
  18. Martínez C, Canals C, Sarina B, Alessandrino EP, Karakasis D, Pulsoni A, et al. Rewlapse of Hodgkin’s Lymphoma after autologous stem cell transplantation (ASCT): Identification of prognostic factors predicting outcome. Blood. (submitted). 
  19. Jabbour E, Hosing C, Ayers G, Nunez R, Anderlini P, Pro B, et al. Pretransplant positive positron emission tomography/gallium scans predict poor outcome in patients with recurrent/refractory Hodgkin lymphoma. Cancer 2007;109:2481-2489 http://dx.doi.org/10.1002/cncr.22714  PMid:17497648 
  20. Moskowitz AJ, Yahalom J, Kewalramani T, Maragulia JC, Vanak JM, Zelenetz AD et al. Pre-transplant functional imaging predicts outcome following autologous stem cell transplant for relapsed and refractory Hodgkin lymphoma. Blood 2010;116:4934-4937 http://dx.doi.org/10.1182/blood-2010-05-282756  PMid:20733154 
  21. Castagna L, Bramanti S, Balzarotti M, Sarina B, Todisco E, Anastasia A, et al. Predictive value of early 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) during salvage chemotherapy in relapsing/refractory Hodgkin Lymphoma (HL) treated with high-dose chemotherapy. Br J Haematol 2009;145:369-372 http://dx.doi.org/10.1111/j.1365-2141.2009.07645.x  PMid:19344403 
  22. Schot BW, Zijlstra JM, Sluiter WJ, van Imhoff G, Pruim J, Vaalburg W, et al. Early FDG-PET assessment in combination with clinical risk scores determines prognosis in recurrent lymphomas. Blood. 2007;109:486-91.  http://dx.doi.org/10.1182/blood-2005-11-006957  PMid:17003382 
  23. Carella AM, Champlin R, Slavin S, McSweeney P, Storb R. Mini-allografts: ongoing trials in humans. Bone Marrow Transplant 2000;25:345-350 http://dx.doi.org/10.1038/sj.bmt.1702204  PMid:10723575 
  24. Younes A, Gopal AK, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, et al. Results of a pivotal phase II study of Brentuximab Vedotin for patients with relapsed or refractory Hodgkin’s Lymphoma. J Clin Oncol. 2012;30:2183-89. http://dx.doi.org/10.1200/JCO.2011.38.0410  PMid:22454421 
  25. Younes A, Sureda A, Ben-Yehuda D, Zinzani PL, Ong TC, Prince M, et al. Panobinostat in patients with relapsed/refractory Hodgkin’s Lymphoma after autologous stem cell transplantation: results of a phase II study. J Clin Oncol. 2012;30:2197-2203. http://dx.doi.org/10.1200/JCO.2011.38.1350  PMid:22547596 
  26. Robinson SP, Goldstone AH, Mackinnon S, Carella AM, Russell N, de Elvira CR, et al. Chemoresistant or aggressive lymphoma predicts for a poor outcome following reduced-intensity allogeneic progenitor cell transplantation: an analysis from the Lymphoma Working Party of the European Group for Blood and Bone Marrow Transplantation. Blood 2002;100:4310-4316 http://dx.doi.org/10.1182/blood-2001-11-0107 PMid:12393626 
  27. Anderlini P, Saliba R, Acholonu S, Okoroji GJ, Donato M, Giralt S, et al. Reduced-intensity allogeneic stem cell transplantationin relapsed and refractory Hodgkin’s disease: lowtransplant-related mortality and impact of intensity of conditioning regimen. Bone Marrow Transplant. 2005;35:943-51. http://dx.doi.org/10.1038/sj.bmt.1704942  PMid:15806128 
  28. Sureda A, Canals C, Arranz R, Caballero MD, Ribera JM, Brune M, et al. Allogeneic stem cell transplantation after reduced-intensity conditioning in patients with relapsed or refractory Hodgkin’s lymphoma: results of the HDR-Allo Study, a prospective clinical trial by the Grupo Espańol de Linfomas/Trasplante de Médula Ósea (GEL/TAMO) and the Lymphoma Working Party of the European Group for Blood and Marrow Transplantation. Leukemia. (submitted). 
  29. Sureda A, Robinson S, Canals C, Carella AM, Boogaerts MA, Caballero D, et al Reduced-intensity conditioning compared with conventional allogeneic stem-cell transplantation in relapsed or refractory Hodgkin’s lymphoma: an analysis from the Lymphoma Working Party of the European Group for Blood and Marrow Transplantation. J Clin Oncol. 2008;26:455-62. http://dx.doi.org/10.1200/JCO.2007.13.2415  PMid:18086796 
  30. Peggs KS, Thomson KJ, Hart DP, Geary J, Morris EC, Yong K, et al. Dose-escalated donor lymphocyte infusions following reduced intensity transplantation: toxicity, chimerism, and disease responses. Blood. 2004;103:1548-56. http://dx.doi.org/10.1182/blood-2003-05-1513  PMid:14576063 
  31. Peggs KS, Thomson KJ, Hart DP, Geary J, Morris EC, Yong K, et al. Dose-escalated donor lymphocyte infusions following reduced intensity transplantation: toxicity, chimerism, and disease responses. Blood. 2004;103:1548-56. http://dx.doi.org/10.1182/blood-2003-05-1513  PMid:14576063 
  32. Hart DP, Avivi I, Thomson KJ, Peggs KS, Morris EC, Goldstone AH, et al. Use of 18F-FDG positron emission tomography following allogeneic transplantation to guide adoptive immunotherapy with donor lymphocyte infusions. Br J Haematol.2005;128:824-829. http://dx.doi.org/10.1111/j.1365-2141.2005.05388.x  PMid:15755287    
  33. Carella AM, Cavaliere M, Lerma E, Ferrara R, Tedeschi L, Romanelli A, et al. Autografting followed by non-myeloablative immunosuppressive chemotherapy and allogeneic peripheral blood hematopoietic stem-cell transplantation as treatment of resistant Hodgkin’s disease and non-Hodgkin’s lymphoma. J Clin Oncol 2000;18:3918-3924. PMid:11099321 
  34. Todisco E, Congiu AG, Castagna L, Nati ST, Santoro A, Carella AM. Tandem autologous/reduced-intensity allograft for relapsed/refractory Hodgkin’s lymphoma: early allotransplant after intensive cytoreduction may maximize graft vs lymphoma effect? Haematologica 2009;94 (s2) abstr. 0090.  
  35. Faulkner RD, Craddock C, Byrne JL, Mahendra P, Haynes AP, Prentice HG, et al. BEAM-alemtuzumab reduced-intensity allogeneic stem cell transplantation for lymphoproliferative diseases: GVHD, toxicity, and survival in 65 patients. Blood. 2004;103:428-34. http://dx.doi.org/10.1182/blood-2003-05-1406  PMid:12969983