Abstract
Corticosteroid refractory graft-versus-host disease (GVHD) is one of the major challenges in the management of allogeneic stem cell transplant recipients. Although numerous agents have been employed to treat this patient population, no standardized second-line therapy exists. In this study, we report our experience with the administration of tocilizumab, an anti-interleukin 6 receptor antibody, in the treatment of steroid refractory GVHD. Tocilizumab was administered to 8 patients with refractory acute (n = 6) or chronic GVHD (cGVHD) (n = 2) once every 3 to 4 weeks. The majority of patients with acute GVHD (aGVHD) had grade IV organ involvement of the skin or gastrointestinal tract, whereas both patients with cGVHD had long-standing severe skin sclerosis at the time of treatment. There were no allergic or infusion-related adverse events. Treatment was discontinued in one patient over concerns that tocilizumab may have worsened preexisting hyperbilirubinemia. Several patients also had transient elevations in serum transaminase values. Infections were the primary adverse events associated with tocilizumab administration. Four patients (67%) with aGVHD had either partial or complete responses apparent within the first 56 days of therapy. One patient with cGVHD had a significant response to therapy, whereas the second had stabilization of disease that allowed for a modest reduction in immune suppressive medications. These results indicate that tocilizumab has activity in the treatment of steroid refractory GVHD and warrants further investigation as a therapeutic option for this disorder.
Keywords: Graft-versus-host disease, Interleukin 6, Tocilizumab, Steroid refractory
INTRODUCTION
Graft-versus-host disease (GVHD) is the major complication associated with allogeneic hematopoietic stem cell transplantation. For several decades, the initial management of acute GVHD (aGVHD) has been the administration of corticosteroids, and approximately 50% of patients will respond to this therapeutic intervention [1,2]. For those patients who fail to respond to steroids, a number of agents have been evaluated as second-line therapy, but no consensus has emerged for the treatment of these patients. Various modalities have been employed to treat steroid refractory GVHD. These agents have generally fallen into broad categories, including cytostatic agents (mycophenolic acid, pentostatin) [3–5], immunomodulating agents (mTor inhibitors, thalidomide, photopheresis) [6–9], and biologic therapies (rituximab, alemtuzumab, infliximab, denileukin difitox) [10–14]. In this second-line treatment setting, response rates have generally been much lower (ie, on the order of 20%–30%), indicating the need for more effective therapies in this patient population.
A prominent characteristic of GVHD is the presence of a proinflammatory milieu that is attributable to conditioning regimen-induced host tissue damage as well as secretion of inflammatory cytokines by alloactivated donor T cells and other immune cell populations [15,16]. This has been the rationale for the administration of agents that interfere with inflammatory cytokine signaling for the therapy of GVHD. Interleukin 6 (IL-6) is a pleiotrophic cytokine that is produced by a variety of cell types and is reportedly elevated in the serum of patients with ongoing GVHD [17]. Moreover, polymorphisms in recipient and donor genotypes that result in increased IL-6 production have been associated with an increase in GVHD severity [18,19]. Recent studies in murine models of GVHD have shown that treatment with an anti-IL-6R antibody significantly reduces GVHD-associated mortality and pathological damage [20,21]. Whether this approach has merit for the treatment of patients with GVHD, however, has not been examined.
Tocilizumab (Actemra™, Roche, Indianapolis, IN) is a humanized anti-IL-6 receptor antibody that blocks IL-6 signaling and has been FDA approved for the treatment of severe active rheumatoid arthritis. It reportedly has remission-inducing efficacy in patients with moderate to severe rheumatoid arthritis, systemic juvenile idiopathic arthritis, and multicentric Castlemans’ disease [22–24]. A recent case report demonstrated that administration of tocilizumab was effective at significantly reducing the severity of GVHD in the gastrointestinal (GI) tract as determined by a marked reduction in the volume of diarrhea [25]. The purpose of this study was therefore to evaluate the efficacy and toxicity of tocilizumab in the treatment of a cohort of patients with steroid refractory GVHD.
PATIENTS, MATERIALS, AND METHODS
Patient Population
Eight patients were treated with off label use of tocilizumab at the discretion of their attending physicians from July 2010 to April 2011. Patients were deemed to be eligible for treatment if they had undergone allogeneic stem cell transplantation and developed steroid refractory, biopsy-proven GVHD, which could be in any tissue site (ie, skin, gut, etc.). Patients were classified as having steroid-resistant aGVHD if any of the following occurred: (1) no change or progression in the stage of skin GVHD after at least 1 week of 2 mg/kg per day or more of methylprednisolone in conjunction with extracorporeal photopheresis (ECP); (2) lack of response of visceral (liver, GI) GVHD despite treatment with 2 mg/kg per day or more of methylprednisolone for at least 72 hours; (3) progression of visceral GVHD despite treatment with 2 mg/kg per day or more of methylprednisolone for at least 48 hours; or (4) visceral GVHD progressing to stage 4 after 24 hours of 2 mg/kg per day or more of methylprednisolone. Patients with protracted acute GVHD who did not respond to a minimum of 0.5 mg/kg/day steroid therapy after 4 weeks were eligible. Finally, patients with classic chronic GVHD (cGVHD) who did not respond within 4 weeks after at least 0.5 mg/kg/day steroid therapy were also eligible. All patients provided written informed consent for hematopoietic progenitor cell transplantation under studies approved by the institutional review board (IRB) at the Medical College of Wisconsin (MCW). GVHD-related data were collected prospectively on each patient. The MCW IRB approved this retrospective analysis of tocilizumab therapy for steroid refractory GVHD.
Treatment
Tocilizumab was administered intravenously at a dose of 8 mg/kg once weekly every 3 to 4 weeks. Patients with documented responses continued to receive treatment on this schedule. Patients who had attained a complete remission were dose reduced to 4 mg/kg as long as they remained on other immune suppressive medications. Tocilizumab was discontinued once patients were able to be taken off all other immune suppressive medications and were free of GVHD for at least 1 month. Patients who did not respond to tocilizumab or had progression of disease were discontinued from therapy.
GVHD and Toxicity Assessment
The extent of aGVHD in GVHD target organs was graded according to the criteria enumerated in Glucksberg et al. [26]. The diagnosis of cGVHD was determined using the National Institutes of Health consensus criteria [27]. Scoring of skin and mucosal abnormalities in patients with cGVHD was based on criteria detailed in the cGVHD response criteria working group report [28]. Toxicity was assessed using the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.
Infection Monitoring
Blood and tissue-directed cultures for bacterial and fungal pathogens (ie, urine, sputum, etc.) were obtained at the discretion of the treating physician when clinically indicated. All patients had weekly cytomegalovirus (CMV) Nucleic Acid Amplified Test (NAAT) testing in the blood as well as Epstein-Barr virus (EBV), human herpes virus 6 (HHV-6), and adenovirus testing by polymerase chain reaction (PCR) in the blood at least every 2 weeks.
Response Definitions
Complete response (CR) was defined as an International Bone Marrow Transplant Registry (IBMTR) score of 0 for the GVHD grading in all evaluable organs. For a response to be scored as CR at day 56 or later, the participant must have been in CR on that day and have had no additional therapy for an earlier progression, partial response (PR), or no response (NR). PR was defined as improvement in 1 or more organs involved with GVHD symptoms without progression in others. For a response to be scored as PR at day 56 or later, the participant must have been in PR on that day and have had no intervening additional therapy for an earlier progression, PR, or NR. Mixed response (MR) was defined as improvement in 1 or more organs with deterioration in another organ manifesting symptoms of GVHD or development of symptoms of GVHD in a new organ. Progression was defined as deterioration in at least 1 organ without any improvement in others. NR was defined as progression with new organ involvement or increased organ-specific symptoms sufficient to increase the organ stage by 1 or more after at least 7 days of study drug administration, or no response (no reduction in any GVHD organ staging) after study drug administration. Patients receiving new additional secondary therapy (including the need to reescalate the steroid dose to ≥2.5 mg/kg/day of prednisone [or a methylprednisolone equivalent of 2 mg/kg/day]) were also classified as nonresponders.
Statistical Analysis
This study is a descriptive case series of 8 consecutive patients treated with tocilizumab for steroid refractory GVHD. Descriptive statistics with a median and a range were used to summarize the number of infusions. GVHD outcomes were described on an individual patient level.
RESULTS
Patient and Disease Characteristics
The patient characteristics are shown in Table 1. For all patients, except 1, this was the initial allogeneic stem cell transplant. Patient UPN 2272, who was transplanted for chronic lymphocytic leukemia (CLL), had failed his first nonmyeloablative transplant with persistent disease and evidence of graft rejection, and was then treated with a second myeloablative transplant from the same unrelated donor. All patients failed therapy with multiple other agents in addition to steroids (Table 2). Organ involvement in patients with acute GVHD consisted of lower GI tract disease in 5 patients (grade II in 1 and grade IV in 4) and skin involvement in 1 patient (grade IV). Two patients had classic cGVHD that was characterized by diffuse sclerosis of the skin with joint contractures.
Table 1.
UPN | Age/Sex | Disease | CMV Serostatus (Donor/Recipient) |
Type of Transplant | Conditioning Regimen | Acute GVHD Prophylaxis |
---|---|---|---|---|---|---|
2031 | 41/M | MM | Neg/Neg | Nonmyeloablative, matched related | TBI 200 cGy | Tacrolimus, MMF |
2090 | 55/M | MM | Neg/Pos | Nonmyeloablative, matched related | TBI 200 cGy | Tacrolimus, MMF |
2200 | 53/M | MM | Neg/Neg | Nonmyeloablative, matched related | TBI 200 cGy | Tacrolimus, MMF |
2272 | 52/M | CLL | Neg/Pos | Myeloablative, matched unrelated | TBI 1200 cGy Cyclophosphamide | Tacrolimus, MMF |
2292 | 52/M | MM | Neg/Pos | Nonmyeloablative, matched unrelated | Fludarabine, TBI 200 cGy | Tacrolimus, MMF, rapamycin, |
2496 | 42/F | AML | Neg/Neg | Myeloablative, mismatched unrelated | Busulfan, cyclophosphamide | ATG, tacrolimus, methotrexate |
2502 | 67/F | CLL | Neg/Neg | Nonmyeloablative, matched unrelated | Fludarabine, TBI 200 cGy | Tacrolimus, MMF |
2503 | 40/F | AML | Pos/Pos | Myeloablative, mismatched unrelated | Busulfan, cyclophosphamide | ATG, tacrolimus, steroids |
MM indicates multiple myeloma; CLL, chronic lymphocytic leukemia; AML, acute myelogenous leukemia; TBI, total body irradiation; MMF, mycophenolate mofetil; ATG, antithymocyte globulin.
Table 2.
UPN | Onset of GVHD (Day Post-BMT) |
Overall Stage of GVHD |
Organ Involvement (Stage)† | GVHD Therapy before Tocilizumab‡ | Tocilizumab Administration (Day Post-BMT) |
|
---|---|---|---|---|---|---|
2031 | 724* | Classic chronic | Skin sclerosis, joint contractures | Steroids, photopheresis, gleevec, plaquinel | 984 | |
2090 | 142 | IV | Gut (IV) | steroids, infliximab (4), campath (2), MMF, budesonide, octreotide | 184 | |
Liver (III) | ||||||
2200 | 487* | Classic chronic | Skin sclerosis, joint contractures | Steroids, photopheresis, infliximab, gleevec, sirolimus, lidocaine, campath (10), rituxan (4), MMF, PUVA, plaquinel | 669 | |
2272 | 26 | IV | Skin (IV) | Steroids, photopheresis | 39 | |
2292 | 106 | II | Gut (II) | Steroids, infliximab (4), budesonide, tacrolimus | 235 | |
2496 | 23 | IV | Skin (II) | Steroids, infliximab (2), budesonide | 51 | |
Gut (IV) | ||||||
2502 | 52 | IV | Skin (II) | Steroids, infliximab (3) | 82 | |
Gut (IV) | ||||||
2503 | 24 | IV | Skin (II) | Steroids, MMF | 32 | |
Gut (IV) |
MMF indicates mycophenolate mofetil; BMT, bone marrow transplant; GVHD, graft-versus-host disease.
Denotes onset of chronic GVHD in these 2 patients.
Gut involvement in these patients was all lower tract disease.
Number in parentheses indicates total number of infusions given of infliximab, campath, or rituxan.
Toxicity and Adverse Events
A total of 31 infusions of tocilizumab were administered during the study period. The median number of infusions was 5 per patient (range: 1–6 infusions), and the median time of administration posttransplantation was 4 months (range: 1–33 months). There was no infusion or allergic-related events. Two patients developed grade 2 elevations in serum transaminases at some time during the course of tocilizumab infusions. Of 31 total infusions, 8 were associated with elevated transaminase levels that were either grade 1 or 2 in severity. One patient (UPN 2031) had a preexisting grade 1 increase in serum aspartate aminotransferase/alanine aminotransferase (ALT/AST) values, but these remained unchanged during all infusions. Tocilizumab was discontinued in 1 patient (UPN 2503) after receiving a single infusion of this agent because of worsening of preexisting grade 1 hyperbilirubinemia. No other patients required discontinuation of this agent because of toxicity.
Infection Assessment
Five patients developed infections during treatment with 13 total documented infections. Nine (69%) of these infectious episodes were of bacterial origin and, except for 1 attributable to Clostridium difficile colitis, were because of bloodstream infections. Two patients developed fungal bloodstream infections attributable to Fusarium sp and Candida sp. Two other patients had viral infections; 1 with CMV colitis, which occurred temporally with grade 4GVHDof the gut, and a second with hemorrhagic cystitis because of BK viruria. CMV viremia was preexisting in 3 patients (UPN 2090, 2272, and 2292) before tocilizumab administration, having developed this condition 1 to 4 weeks before antibody treatment. Two patients cleared viremia after starting tocilizumab (4 months and 2 months, respectively). The third patient died 9 days after initiation of treatment with detectable CMV viremia without evidence of CMV disease. No patient developed CMV viremia after initiation of tocilizumab. None of these patients had evidence of EBV, HHV-6, or adenovirus reactivation or disease while on therapy.
Response to Treatment with Tocilizumab
The median time from onset of GVHD to administration of tocilizumab was 36 days (range: 8–260 days). An overall response was observed in 4 patients (67%) with aGVHD (Table 3). Two of 6 patients died with grade 4 aGVHD of the lower gastrointestinal tract. One patient had no demonstrable response to tocilizumab, whereas the second died 9 days after the first dose of tocilizumab with active GVHD and was deemed to be nonevaluable. Two patients had a complete response at day 56, whereas 2 others had partial responses. The complete responses were characterized by complete resolution of diarrhea. In the responding group of patients, immune suppressive medications were reduced in all patients to varying degrees (Table 3).
Table 3.
UPN | Response (Day 56) | Current Status | Cause of Death | Pre-Tocilizumab Therapy | Post-Tocilizumab Therapy† |
---|---|---|---|---|---|
2031 | Stabilization of skin sclerosis | Alive, d+1123 | — | Prednisone 30 mg every day, gleevec, rapamycin, photopheresis | Prednisone 20/10 every other day, rapamycin, photopheresis, plaquenil |
2090 | NE | Dead, d+195 | GVHD | NA | NA |
2200 | PR | Alive, d+752 | — | Prednisone 10 mg every day, rapamycin, gleevec, lidocaine, photopheresis every 2 weeks | Prednisone 10 mg every other day, photopheresis every 6 weeks |
2272 | PR | Dead, d+213 | Infection | SoluMedrol 65 mg twice a day, tacrolimus | Prednisone 30 mg every day |
2292‡ | CR | Alive, d+496 | — | SoluMedrol 10 mg every day, tacrolimus, MMF, budesonide | Hydrocortisone 20/10 mg every day, budesonide |
2496 | CR | Alive, d+174 | — | SoluMedrol 50 mg every day, MMF, budesonide | Prednisone 20 mg every day, budesonide |
2502 | PR | Alive, d+157 | — | SoluMedrol 50 mg twice a day, MMF 1 g twice a day, tacrolimus | SoluMedrol 30 mg every day, MMF 750 mg twice a day, tacrolimus |
2503 | NR | Dead, d+85 | GVHD | NA | NA |
NE indicates not evaluable; CR, complete response; PR, partial response; NR, no response; NA, not applicable; GVHD, graft-versus-host disease.
Immune suppressive therapy pre- and post-tocilizumab is shown for responding patients only.
Post-tocilizumab therapy refers to the immune suppressive regimen that each patient was taking at the time of last contact.
Post-tocilizumab steroid dose in this patient was replacement therapy to prevent adrenal insufficiency.
Two patients (UPN 2031 and 2200) who were treated with tocilizumab had classic cGVHD, which was manifested predominantly by sclerosis of the skin that was long-standing and refractory to multiple prior immune suppressive medications (Table 2). No validated scoring system has been identified for the assessment of patients with sclerotic skin changes during cGVHD [29]. For that reason, we employed serial measurements of joint mobility as a surrogate indicator for skin sclerosis and also incorporated elements of the NIH consensus criteria for other aspects of skin GVHD [29]. One patient had stabilization of disease while taking tocilizumab. He had no alteration in his joint range of motion when serially evaluated by a physical therapist and no improvement noted on a cGVHD assessment scale. The steroid dose, however, was modestly reduced (Table 3). The second patient (UPN 2200) had a significant response to tocilizumab. The most severely affected area of skin sclerosis and joint contractures were in the lower extremities for this patient. The clinical response in this patient was characterized by measureable improvements in the range of motion in multiple joints, primarily in hip extension, knee flexion and extension, and ankle dorsiflexion (Supplementary Table 1). Increased flexibility in his lower extremities allowed for improved gait, which resulted in a significant augmentation in ambulation distance. Furthermore, there was improvement in overall skin and mucosal scores as well as a substantial reduction in his overall level of immune suppression.
DISCUSSION
In this report, we examined the efficacy of tocilizumab, an IL-6 receptor antibody that blocks signaling of IL-6, in the treatment of patients with steroid refractory GVHD. We observed overall clinical responses in 5 of 8 patients treated with tocilizumab. The majority of patients with aGVHD had overall grade 4 disease, with the GI tract being the predominant target organ. The median time from onset of GVHD to administration of tocilizumab was 29 days in this group. Thus, most patients who had failed steroids had persistent, severe GVHD for several weeks before tocilizumab was added to the regimen. Tocilizumab also appeared to have some efficacy in the treatment of skin sclerosis because of cGVHD. Both patients treated with this agent had failed multiple prior modalities and had significant functional limitations because of concurrent joint contractures. One patient had stabilization of disease, which was associated with a modest reduction in his steroid dose, whereas the second patient had a more dramatic response with objective improvement in joint mobility, a reduction in the amount of skin involvement, and substantial decrease in immune suppressive medications.
The rationale for the administration of tocilizumab derived from preclinical studies that had demonstrated that blockade of IL-6 signaling attenuates the severity of GVHD [20,21]. Protection from GVHD was attributed to a recalibration of the effector and regulatory arms of the immune system. This was characterized by a reduction in proinflammatory TH1 and TH17 cells with a commensurate increase in the number of regulatory T cells [20]. In particular, these preclinical studies demonstrated a role for IL-6 in mediating pathologic damage within the colon microenvironment as increased levels of IL-6 were also detected within the colon microenvironment [29]. Furthermore, blockade of IL-6 signaling with an anti-IL-6R antibody resulted in more profound protection in the colon relative to other GVHD target organs, such as the liver and lung. Thus, the clinical responses we observed in the gut in tocilizumab treated patients, coupled with a prior report [25], support a role for IL-6 in the pathophysiology of GI GVHD. Whether blockade of IL-6 signaling in allogeneic transplant recipients is able to augment reconstitution of regulatory T cells, as observed in murine studies, was not addressed in this study and will require further investigation.
The responsiveness of sclerotic skin manifestations in 1 patient treated with tocilizumab may provide some new insight into the pathophysiology of skin fibrosis during GVHD. Recent studies from Hill and colleagues [30] have demonstrated that cGVHD induced in mice after transplantation of granulocyte-colony stimulating factor (G-CSF)-mobilized stem cell grafts is associated with development of scleroderma in the skin. This was reportedly because of the overproduction of IL-17. IL-6 reportedly plays a pivotal role in the differentiation of TH17 cells from naive T cells. Naive T cells stimulated with anti-CD3 and anti-CD28 antibodies in the presence of transforming growth factor-beta (TGF-β) differentiate into induced regulatory T cells, whereas exposure to IL-6 along with TGF-β drives them to become TH17 cells capable of secreting IL-17 [31,32]. Thus, blockade of IL-6 may serve to decrease differentiation of naïve T cells into TH17 cells and thereby attenuate skin sclerosis. Alternatively, reported effects of tocilizumab on the memory B cell compartment [33] is another mechanism by which this agent may have facilitated a response, given the role that B cells play in cGVHD [13]. Notably, anti-IL-6 therapy reportedly suppresses procollagen production in fibroblasts from scleroderma patients [34]. A recent report has also demonstrated alleviation of fibrotic skin changes in a small group of patients with primary scleroderma treated with tocilizumab [35]. Collectively, these data point to a role for IL-6 in the fibrosis and sclerotic skin changes that can occur as a consequence of cGVHD, and suggest that blockade of IL-6 may represent a novel treatment approach for this complication.
Adverse events associated with tocilizumab administration were consistent with those previously reported for this agent [22,23,36,37] and those typically observed in allogeneic transplant recipients (eg, infections). There were elevations in serum transaminases in several patients, but these tended to be modest and did not increase with subsequent infusions. One patient required a liver biopsy because of hyperbilirubinemia, which worsened to grade 4 after initiation of tocilizumab. The biopsy showed no evidence of GVHD or CMV hepatitis, with only cholestasis noted on pathologic examination. The cause of her hyperbilirubinemia remained undetermined, and it was uncertain the extent to which tocilizumab may have contributed to this abnormality. Infections were the major concurrent event observed in many of these patients. It was difficult, however, to determine whether tocilizumab may have predisposed patients to infectious complications because all of these patients had recently received multiple modalities for their GVHD treatment and were all still taking other immune suppressive medications at the time of tocilizumab administration. All patients who were seropositive for CMV reactivated this virus posttransplantation. However, 3 of these 4 patients had CMV viremia that antedated tocilizumab administration. One died within 10 days after tocilizumab was begun with refractory GVHD. In the remaining 2 patients, viremia persisted for several months despite continuous anti-CMV therapy, but none developed CMV disease and eventually viremia cleared in both patients. The fourth patient developed CMV colitis post-tocilizumab administration. The remaining infections that were observed in this cohort of patients were either bacterial or fungal in origin.
The tocilizumab dose administered in this study was based on prior experience derived from the use of this agent in adult patients with rheumatoid arthritis [22]. In that report, a dose of 8 mg/kg was found to be superior to 4 mg/kg when assessed at 3 months. For that reason, all patients were initially treated at that dose, although responding patients who remained on immune suppressive medications were dose reduced to 4 mg/kg in an effort to minimize exposure to another immunosuppressive agent. We also employed an administration schedule that was derived from prior clinical arthritis studies where tocilizumab was given approximately every 4 weeks. Although we were able to document clinical responses in patients with steroid-refractory GVHD, we do not know if this is the optimal schedule. Prior studies have shown that intravenous innunoglobulin has a much shorter half-life in bone marrow transplant recipients when compared with normal controls [38]. Thus, it is possible that a more frequent administration schedule might be more clinically efficacious. In that regard, a recent study in patients with Crohn’s disease, where tocilizumab administration showed remission-inducing activity, was given on an every 2 week basis [39]. Further studies will be needed to define this question.
In summary, these results demonstrate that tocilizumab has activity in the treatment of steroid refractory GVHD. The response observed in patients with lower GI tract involvement coupled with preclinical studies showing protection from GVHD within the colon microenvironment suggests that blockade of IL-6 signaling may represent a novel approach for the prevention and/or treatment of GVHD within this tissue site. Additionally, tocilizumab may also provide benefits in the therapy of skin sclerosis that occurs as a consequence of cGVHD. Tocilizumab was generally well tolerated, although whether this therapy predisposes patients to opportunistic infections remains to be determined. We conclude that further investigation of this agent in patients with both aGVHD and cGVHD is warranted.
Supplementary Material
Footnotes
Financial disclosure: The authors have nothing to disclose.
SUPPLEMENTARY DATA
Supplementary data related to this article can be found online at doi:10.1016/j.bbmt.2011.07.001.
REFERENCES
- 1.Weisdorf D, Haake R, Blazar B, et al. Treatment of moderate/severe acute graft versus host disease after allogeneic bone marrow transplantation: an analysis of clinical risk features and outcome. Blood. 1990;75:1024–1030. [PubMed] [Google Scholar]
- 2.Macmillan ML, Weisdorf DJ, Wagner JE, et al. Response of 443 patients to steroids as primary therapy for acute graft versus host disease: comparison of grading systems. Biol Blood Marrow Transplant. 2002;8:387–394. doi: 10.1053/bbmt.2002.v8.pm12171485. [DOI] [PubMed] [Google Scholar]
- 3.Basara N, Blau WI, Romer E, et al. Mycophenolate mofetil for the treatment of acute and chronic GVHD in bone marrow transplant patients. Bone Marrow Transplant. 1998;22:61–65. doi: 10.1038/sj.bmt.1701281. [DOI] [PubMed] [Google Scholar]
- 4.Kim JG, Sohn SK, Kim DH, et al. Different efficacy of mycophenolate mofetil as salvage treatment fro acute and chronic GVHD after allogeneic stem cell transplant. Eur J Haematol. 2004;73:56–61. doi: 10.1111/j.1600-0609.2004.00247.x. [DOI] [PubMed] [Google Scholar]
- 5.Bolanos-Meade J, Jacobsohn DA, Margolis J, et al. Pentostatin in steroid-refractory acute graft versus host disease. J Clin Oncol. 2005;23:2661–2668. doi: 10.1200/JCO.2005.06.130. [DOI] [PubMed] [Google Scholar]
- 6.Benito AI, Furlong T, Martin PJ, et al. Sirolimus (Rapamycin) for the treatment of steroid-refractory acute graft versus host disease. Transplantation. 2001;72:1924–1929. doi: 10.1097/00007890-200112270-00010. [DOI] [PubMed] [Google Scholar]
- 7.Hoda D, Pidala J, Salgado-Vila N, et al. Sirolimus for treatment of steroid-refractory acute graft versus host disease. Bone Marrow Transplant. 2010;45:1347–1351. doi: 10.1038/bmt.2009.343. [DOI] [PubMed] [Google Scholar]
- 8.Vogelsang GB, Hess AD, Santos GW. Thalidomide for treatment of graft versus host disease. Bone Marrow Transplant. 1988;3:393–398. [PubMed] [Google Scholar]
- 9.Greinix HT, Volc-Platzer B, Kalhs P, et al. Extracorporeal photochemotherapy in the treatment of severe steroid-refractory acute graft versus host disease: a pilot study. Blood. 2000;96:2426–2431. [PubMed] [Google Scholar]
- 10.Couriel D, Saliba R, Hicks K, et al. Tumor necrosis alpha blockade for the treatment of acute GVHD. Blood. 2004;104:649–654. doi: 10.1182/blood-2003-12-4241. [DOI] [PubMed] [Google Scholar]
- 11.Ho VT, Zahrieh D, Hochberg E, et al. Safety and efficacy of denileukin diftitox in patients with steroid-refractory acute graft versus host disease after allogeneic hematopoietic stem cell transplantation. Blood. 2004;104:1224–1226. doi: 10.1182/blood-2004-01-0028. [DOI] [PubMed] [Google Scholar]
- 12.Shaughnessy PJ, Bachier C, Grimley M, et al. Denileukin difitox for the treatment of steroid-resistant acute graft versus host disease. Biol Blood Marrow Transplant. 2005;11:188–193. doi: 10.1016/j.bbmt.2004.11.022. [DOI] [PubMed] [Google Scholar]
- 13.Cutler C, Miklos D, Kim HT, et al. Rituximab for steroid-refractory chronic graft versus host disease. Blood. 2006;108:756–762. doi: 10.1182/blood-2006-01-0233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Schub N, Gunther A, Schrauder A, et al. Therapy of steroid-refractory acute GVHD with CD52 antibody alemtuzumab is effective. Bone Marrow Transplant. 2011;46:143–147. doi: 10.1038/bmt.2010.68. [DOI] [PubMed] [Google Scholar]
- 15.Hill GR, Crawford JM, Cooke KR, Brinson YS, Pan L, Ferrara JL. Total body irradiation and acute graft versus host disease: the role of gastrointestinal damage and inflammatory cytokines. Blood. 1997;90:3204–3213. [PubMed] [Google Scholar]
- 16.Ferrara JL, Levine JE, Reddy P, Holler E. Graft versus host disease. Lancet. 2009;373:1550–1561. doi: 10.1016/S0140-6736(09)60237-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Imamura M, Hashino S, Kobayashi H, et al. Serum cytokine levels in bone marrow transplantation: synergistic interaction of interleukin 6, interferon gamma, and tumor necrosis factor alpha in graft versus host disease. Bone Marrow Transplant. 1994;13:745–751. [PubMed] [Google Scholar]
- 18.Cavet J, Dickinson AM, Norden J, Taylor PRA, Jackson GH, Middleton PG. Interferon γ and interleukin 6 gene polymorphisms associate with graft versus host disease in HLA-matched sibling bone marrow transplantation. Blood. 2001;98:1594–1600. doi: 10.1182/blood.v98.5.1594. [DOI] [PubMed] [Google Scholar]
- 19.Socie G, Loiseau P, Tamouza R, et al. Both genetic and clinical factors predict the development of graft versus host disease after allogeneic hematopoietic stem cell transplantation. Transplantation. 2001;72:699–706. doi: 10.1097/00007890-200108270-00024. [DOI] [PubMed] [Google Scholar]
- 20.Chen X, Das R, Komorowski R, Beres A, Hessner MJ, Mihara M, Drobyski WR. Blockade of interleukin 6 signaling augments regulatory T cell reconstitution and attenuates the severity of graft versus host disease. Blood. 2009;114:891–900. doi: 10.1182/blood-2009-01-197178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tawara I, Koyama M, Liu C, et al. Interleukin 6 modulates graft versus host disease after experimental allogeneic bone marrow transplantation. Clin Cancer Res. 2011;17:77–88. doi: 10.1158/1078-0432.CCR-10-1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Nishimoto N, Yoshizaki K, Miyasaka N, et al. Treatment of rheumatoid arthritis with humanized anti-interleukin 6 receptor antibody. Arthritis Rheum. 2004;50:1761–1769. doi: 10.1002/art.20303. [DOI] [PubMed] [Google Scholar]
- 23.Yokota S, Imagawa T, Mori M, et al. Efficacy and safety of tocilizumab in patients with systemic-onset juvenile idiopathic arthritis: a randomized, double-blind, placebo-controlled, withdrawal phase III trial. Lancet. 2008;371:998–1006. doi: 10.1016/S0140-6736(08)60454-7. [DOI] [PubMed] [Google Scholar]
- 24.Matsuyama M, Suzuki T, Tsuboi H, et al. Anti-interleukin 6 receptor antibody (Tocilizumab) treatment of multicentric Castleman’s disease. Intern Med. 2007;46:771–774. doi: 10.2169/internalmedicine.46.6262. [DOI] [PubMed] [Google Scholar]
- 25.Gergis U, Arnason J, Yantiss R, Shore T, Wissa U, Feldman E. Effectiveness and safety of tocilizumab, an anti-interleukin 6 receptor monoclonal antibody in a patient with refractory GI graft versus host disease. J Clin Oncol. 2010;28:e602–e604. doi: 10.1200/JCO.2010.29.1682. [DOI] [PubMed] [Google Scholar]
- 26.Glucksberg H, Storb R, Fefer A, et al. Clinical manifestations of graft-versus-host disease in human recipients of marrow from HLA matched sibling donors. Transplantation. 1974;18:295–304. doi: 10.1097/00007890-197410000-00001. [DOI] [PubMed] [Google Scholar]
- 27.Filipovich AH, Weisdorf D, Pavletic S, et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft versus host disease: I. Diagnosis and staging working group report. Biol Blood Marrow Transplant. 2005;11:945–956. doi: 10.1016/j.bbmt.2005.09.004. [DOI] [PubMed] [Google Scholar]
- 28.Pavletic SZ, Martin P, Lee SJ, et al. Measuring therapeutic response in chronic graft versus host disease: National Institutes of Health Consensus Development Project on criteria for clinical trials in chronic graft versus host disease: IV. Response criteria working group report. Biol Blood Marrow Transplant. 2006;12:252–266. doi: 10.1016/j.bbmt.2006.01.008. [DOI] [PubMed] [Google Scholar]
- 29.Das R, Chen X, Komorowski R, Hessner MJ, Drobyski WR. Interleukin 23 secretion by donor antigen presenting cells is critical for organ-specific pathology in graft versus host disease. Blood. 2009;113:2352–2362. doi: 10.1182/blood-2008-08-175448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hill GR, Olver SD, Kuns RD, et al. Stem cell mobilization with G-CSF induces type 17 differentiation and promotes scleroderma. Blood. 2010;116:819–828. doi: 10.1182/blood-2009-11-256495. [DOI] [PubMed] [Google Scholar]
- 31.Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–238. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
- 32.Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGF beta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17 producing T cells. Immunity. 2006;24:179–186. doi: 10.1016/j.immuni.2006.01.001. [DOI] [PubMed] [Google Scholar]
- 33.Roll P, Muhammad K, Schumann M, Kleinert S, Einsele H, Dorner T, Tony HO. In vivo effect of the anti-interleukin 6 receptor inhibitor tocilizumab on the B cell compartment. Arthritis Rheum. 2011 May;63(5):1255–1264. doi: 10.1002/art.30242. [DOI] [PubMed] [Google Scholar]
- 34.Kawaguchi Y, Hara M, Wright TM. Endogenous IL-1 alpha from systemic sclerosis fibroblasts induces IL-6 and PDGF-A. J Clin Invest. 1999;103:1253–1260. doi: 10.1172/JCI4304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Shima Y, Kuwahara Y, Murota H, et al. The skin of patients with systemic sclerosis softened during treatment with anti-IL receptor antibody tocilizumab. Rheumatology. 2010;49:2408–2412. doi: 10.1093/rheumatology/keq275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Nishimoto N, Miyasaka N, Yamamoto K, Kawai S, Takeuchi T, Azuma J. Long term safety and efficacy of tocilizumab, an anit-IL-6-receptor monoclonal antibody, in monotherapy, in patients with rheumatoid arthritis (the STREAM study): evidence of safety and efficacy in a 5 year extension study. Ann Rheum Dis. 2009;68:1580–1584. doi: 10.1136/ard.2008.092866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Campbell L, Chen C, Bhagat SS, Parker RA, Ostor AJK. Risk of adverse events including serious infections in rheumatoid arthritis patients treated with tocilizumab; a systematic literature review and meta-analysis of randomized controlled trials. Rheumatology. 2011;50:552–562. doi: 10.1093/rheumatology/keq343. [DOI] [PubMed] [Google Scholar]
- 38.Rand KH, Gibbs K, Derendorf H, Graham-Pole J. Pharmacokinetics of intravenous immunoglobulin (Gammagard) in bone marrow transplant patients. J Clin Pharmacol. 1991;31:1151–1154. doi: 10.1002/j.1552-4604.1991.tb03688.x. [DOI] [PubMed] [Google Scholar]
- 39.Ito H, Takazoe M, Fukuda Y, et al. A pilot randomized trial of a human anti-IL-6 receptor monoclonal antibody in active Crohn’s disease. Gastroenterology. 2004;126:989–996. doi: 10.1053/j.gastro.2004.01.012. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.