Next Article in Journal
A High-Content Screen for the Identification of Plant Extracts with Insulin Secretion-Modulating Activity
Next Article in Special Issue
Chondroitin Sulfate and Fucosylated Chondroitin Sulfate as Stimulators of Hematopoiesis in Cyclophosphamide-Induced Mice
Previous Article in Journal
Lung Fibrosis after COVID-19: Treatment Prospects
Previous Article in Special Issue
Relationship between CYP3A5 Polymorphism and Tacrolimus Blood Concentration Changes in Allogeneic Hematopoietic Stem Cell Transplant Recipients during Continuous Infusion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Extracorporeal Photopheresis in Children with Chronic Graft-Versus-Host Disease

by
Andrey Kozlov
*,
Maria Estrina
,
Olesia Paina
,
Tatiana Bykova
,
Anna Osipova
,
Polina Kozhokar
,
Zhemal Rakhmanova
,
Irina Solodova
,
Elena Morozova
,
Alexander Alyansky
,
Irina Kulagina
,
Asmik Gevorgian
,
Anna Dotsenko
,
Ivan Moiseev
,
Alexey Chukhlovin
,
Alexander Kulagin
,
Sergey Bondarenko
,
Elena Semenova
and
Ludmila Zubarovskaya
RM Gorbacheva Research Institute, Pavlov University, 197022 St. Petersburg, Russia
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2021, 14(8), 808; https://doi.org/10.3390/ph14080808
Submission received: 21 June 2021 / Revised: 6 August 2021 / Accepted: 13 August 2021 / Published: 17 August 2021
(This article belongs to the Special Issue Pharmaceutical and Cellular Strategies to Treat GVHD)

Abstract

:
Chronic graft versus host disease (cGVHD) remains a major complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). It significantly decreases survival and quality of life. The present study demonstrates retrospective data on extracorporeal photopheresis (ECP) in children with cGVHD. A total of 42 children with steroid-refractory cGVHD were enrolled in the study. The majority of patients had acute leukemia (n = 32, 76%). All patients received ECP as second (n = 18, 43%) or third (n = 24, 57%) line of therapy. Initial ECP schedule consisted of bimonthly regimen for two consecutive days with possibility of further tapering according to response. Any concurrent treatment administered before ECP could be continued if considered necessary. Complete response to ECP was registered in seven (17%) patients and partial response in 24 (57%). Overall response according to organ involvement was as follows: skin (n = 24, 75%), mucous membranes (n = 16, 73%), liver (n = 8, 80%), gut (n = 4, 80%), lungs (n = 2, 22%) and joints (n = 2, 67%). Five-year overall, progression-free and failure-free survival was 57%, 56% and 30%, respectively. Non-relapse mortality at 5 years was 14%. We didn’t observe any clinically significant complications in children that could be attributed to the procedure. ECP remains important and safe treatment option in children with cGVHD.

1. Introduction

Chronic graft-versus-host disease (cGVHD) remains a major complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). This disorder may significantly decrease survival and quality of life, despite efficient preventive and therapeutic options. Non-relapse mortality (NRM) in surviving patients with cGVHD is increased within decades after allo-HSCT [1]. More recently, posttransplant cyclophosphamide (PtCy) and ruxolitinib became game changers that improved clinical outcome [2,3,4,5]. These innovative approaches were readily implemented into routine practice, thus resulting in lower cGVHD incidence and excellent control of cGVHD symptoms in most patients. Autoimmune-like symptoms are characteristic to cGVHD, suggesting a multimodal pathogenesis which is currently poorly understood, with immune dysregulation being the leading mechanism [6,7,8]. Resulting immune-mediated inflammatory reactions and connective tissue fibrosis comprise the pathophysiology of cGVHD [5].
Treatment of cGVHDutilizes immunosupressive and/or immunomodulatory therapies. It is always a challenge to treat a disorder with unclear and complex pathogenesis since exact mechanisms of distinct curative approaches are not well defined. With emergence of new treatment options, the role of previously established therapies in children should be reassessed, including extracorporeal photopheresis (ECP). Precise biological action of ECP is unknown, but apoptosis of pathogenic T-cells is supposed to be the most significant factor. Tolerogenic dendritic cells cause switch of immune response profile, whereas increase in the number of T-regulatory cells and natural killer cells are also probable effects of ECP [9].
Existing information on ECP usage in pediatrics is limited to pilot studies. But, as in other disorders, cGVHD may have specific pediatric features compared to adults, with distinct patterns of clinical response. These circumstances necessitate analysis of ECP effectiveness in children separately from adults. ECP is used for cGVHD treatment for approximately 25 years [10]. Multiple pilot studies demonstrated its efficiency in steroid-refractory (SR)cGVHD [11]. However, only few randomized trials were performed, with major limitations for the study design, thus precluding definite evaluation of ECP effects in cGVHD management [12,13]. The aim of present study was to summarize retrospective data from our clinics on ECP efficiency in children with SR cGVHD.

2. Results

Patients characteristics are listed in Table 1.
Complete response to ECP-based therapy was registered in seven (17%) patients and partial response in 24 (57%). Overall response (ORR) according to organ involvement was as follows: skin (n = 24, 75%), mucous membranes (n = 16, 73%), liver (n = 8, 80%), gut (n = 4, 80%), lungs (n = 2, 22%) and joints (n = 2, 67%) (Table 2).
Prior to ECP administration median dosage of steroids was 0.5 mg/kg/day (range 0–1) and after treatment with ECP median dosage was 0.25 mg/kg/day (range 0–2) (p = 0.0003). Overall steroids were tapered in 25 children (66%). Non-relapse mortality (NRM) after 5 years of observation was 14% (CI95%, 5–28%) and cumulative incidence of relapse (CIR), 23% (CI95%, 11–38%).Relapse of the malignancy (n = 11) was the main cause of death, and only four patients succumbed to cGVHD. With median follow-up of 774 days (180–4541), OS and PFS at 5 years were 57% (CI95%, 39–72%) and 56% (CI95%, 37–72%), respectively. FFS at 5 years was 30% (CI95% 16–46%) (Figure 1, Figure 2 and Figure 3).
Eighteen patients (43%) required correction of IST after ECP treatment, due to insufficient clinical effect. Only response to ECP was associated with improved OS (p = 0.0002). No other potential prognostic factors affected survival in the study (Table 3). Multivariate analysis was not performed as only one factor was statistically significantly associated with survival.
We didn’t observe any clinically significant complications attributable to ECP. The most common adverse effects were catheter-associated (n = 5) which did not result into the ECP discontinuation. Three patients had recurrent venous thrombosis, and catheter-associated infections were detected in two cases. One patient developed a severe blood electrolyte imbalance that was successfully corrected. Among eight children that received combination of ECP with ruxolitinib one episode (13%) of transient cytopenia was registered. It did not result in discontinuation or correction of therapy. Other well-known complications of ECP, such as arterial hypotension, decrease in peripheral blood counts, pyrexia and abdominal pain syndrome were uncommon and mild and were not thoroughly recorded in this study.

3. Discussion

Severe cGVHD, solid organ transplantation and cutaneous T-cell lymphoma are approved indications for ECP. Any other indications, including various autoimmune disorders, have low strength of recommendation [11,14]. Probably, unique mechanism of ECP action has immunological consequence only in the three abovementioned disorders.
Less than 20% of patients with cGVHD adequately respond to initial therapy, thus presuming steroid resistance to be a common clinical situation [15]. Hence, the majority of patients with cGVHD would require treatment in addition to steroid therapy. Though mild cGVHD can be often managed with local therapies, systemic treatment is also a rational option in rare cases, according to established criteria [16]. In the present ECP study, several children with mild cGVHD did not significantly influence overall data analysis, due to low number of such patients.
The reported data on long-term outcomes in children with SR cGVHD are limited. Therefore, emergence of new treatment strategies requires regular re-evaluation of optimal management [1]. All existing anti-GVHD drugs, or non-pharmacological methods show a significant rate of treatment failure [4,17]. Treating physician needs a wide range of therapeutic options to perform comprehensive cGVHD management. A significant proportion of children (43%) in our study also required further correction of IST after ECP, due to inadequate or transient clinical response. Hence, several equally effective treatment options should be available in order to manage cGVHD patients. In most instances, only combination therapy may provide control of SR cGVHD [18]. Another interesting and yet poorly studied question is the order of administration of immunosuppressive therapies for SR cGVHD. It seems that this order may play a substantial role in immune tolerance induction.
According to data reported elsewhere, ORR and OS in patients with SR cGVHD after ECP exceed 60% [19]. Our study demonstrated similar results, i.e., a high response rate to ECP-based therapy in all the organs, except of lungs. When discussing the response to ECP one should assume that patients always receive combination IST with ECP being only a component of therapeutic approach, so whether there is a genuine response to ECP or response to combination IST remains unanswered. Steroid-sparing effect of ECP-based therapy was demonstrated in our study with median dosage of steroids of 0.5 mg/kg prior to treatment and 0.25 mg/kg after the therapy (p = 0.0003). This important benefit of ECP reduces adverse effects associated with prolonged steroid therapy. Chronic GVHD in children could theoretically have some specific features compared to adults, like in other diseases. This opportunity is supported by different immune reconstitution features in children and adults [20]. But we didn’t observe any differences in response rate and survival in children with SR cGVHD compared to adults, in terms of ECP effectiveness. Similar data had been registered earlier in pediatric population [21].
Despite established criteria for cGVHD response, assessment of this important outcome measure is still inaccurate, due to subjective clinical interpretations, especially in retrospective analysis. OS is probably the most important and objective endpoint. In the present study, long-term OS (57%) and PFS (56%) were relatively high for this unfavorable group which predominantly comprised of high-risk patients. Relatively low level of CIR (23%) demonstrates fundamental opportunity of ECP to control SR cGVHD, without interfering in graft-versus-tumor effect [22,23,24]. The present study seems to be the first to demonstrate FFS in pediatric population treated with ECP. This important outcome measurement demonstrates more complex characteristics of therapy efficiency as compared to simple response to ECP. In our study, the failure-free survival rate was 30%. It means that only one-third of the children can be cured from cGVHD after ECP. The majority of patients, despite ECP, would require additional IST, due to persistence of clinically significant cGVHD symptoms. Anyway, ECP remains established and safe treatment options with only uncommon catheter-associated complications and absence of clinically significant systemic adverse effects. These data are in accordance with previously published results [25,26]. Chronic GVHD is usually characterized by low incidence of CR, despite the type of applied therapy. Therefore, various treatment modes are used in order to achieve the best response. The same patterns of response in cGVHD and classical scleroderma suggest common pathogenetic features of both conditions [27].
ECP in children is associated with several patient-specific parameters [28]. Low weight is among most significant factors that prevent classical ECP in children < 8 kg, due to their inability to tolerate this extracorporeal procedure. Low-weight patients could be managed by means of small-scale ECP technique if not eligible for apheresis [29]. Hence, ECP can be performed in any patient regardless of weight.
Some investigators demonstrated improved OS in responders to ECP compared to non-responders [30]. Such approach seems to be somewhat speculative, since the group characteristics are not specified in details and the patients from these two groups may differ in their fundamental characteristics. Moreover, treatment of non-responders to ECP has changed nowadays, due to introduction of novel effective drugs, e.g., ruxolitinib. Non-responders could be also refractory to any IST applied (not necessary ECP), for some intrinsic biological reasons not associated with treatment. This biased design strategy prevents direct comparison of the two groups differing in response to ECP.
Nevertheless, improved OS in our study was associated only with clinical response to ECP. These data support fundamental need for cGVHD control in allo-HSCT survivors. All other patient and transplant characteristics didn’t impact survival, probably, due to limited number of enrolled patients. Addition of ruxolitinib treatment to ECP also didn’t result in improved survival. These data can’t replace direct comparisons between ECP and ruxolitinib therapy, since the patients subjected to combination therapy exhibit more resistant course of cGVHD. Further studies to define most optimal cGVHD strategies and their application order are quite reasonable. In our study, ECP was safely co-administered with ruxolitinib in eight children (19%). This is in concordance with recently published data and proves a fundamental opportunity to combine these treatment options, if required [18].
Durable response is the main goal of the GVHD therapy, and this effect may be achieved only after immune tolerance induction. Generally, cGVHD prophylaxis should lead to tolerance between donor and recipient cells by ca. 3 months after allo-HSCT [7]. In case of cGVHD, tolerance cannot be achieved after standard drug prophylaxis, or this balance may be impaired. ECP results not only in cGVHD symptoms amelioration, but it also provides extra time for development of physiological tolerance. Better understanding of immune tolerance induction in patients with cGVHD is important since it would lead to the appearance of new treatment paradigms. According to prolonged FFS observed in the present study, ECP-based therapy induces continuous immune tolerance in, approximately, one-third of children with SRcGVHD. In general, even after ECP-failure, cGVHD was well controlled, i.e., only four patients in the study succumbed due to cGVHD. This fact hypothesizes that ECP can make immune system more sensitive for further therapy. Long-term NRM was 14%, comparable to previously published data [1]. This level should be regarded as high, and additional, more effective treatment options are needed for cGVHD control in non-responders to ECP. Ruxolitinib and ibrutinib are among the most promising agents in these clinical situations.

4. Materials and Methods

The study was performed in RM Gorbacheva Research Institute at the I.Pavlov University (St. Petersburg, Russian Federation). A total of 42 children with SR cGVHD were enrolled. Median age was 13 years (1–18). There were 25 (59%) males and 17 (41%) females in the study. The majority of patients had acute leukemia (n = 32, 76%), other diagnoses included myelodisplastic syndrome/juvenile myelomonocytic leukemia (MDS/JMML) (n = 3, 7%), Fanconi anemia (n = 2, 5%), chronic myelogenous leukemia (n = 2, 5%), Hodgkin’s disease (n = 2, 5%) and Hurler syndrome (n = 1, 2%). Patients were attributed to the high-risk group (n = 30, 71%) in case of acute leukemia (≥2 remissions) and MDS/JMML, all other patients were classified to standard risk group prior to allo-HSCT. GVHD prophylaxis was mostly based on the post-transplant cyclophosphamide (PtCy) (n = 18, 43%) and anti-thymocyte globuline (n = 18, 43%). HLA-matched related donors were used as stem cell source in seven patients (17%); haploidentical donors, in 17 patients (40%); HLA-matched unrelated donors, in 13 (31%), and mismatched unrelated donors, in five cases (12%). Myeloablative and reduced-intensity conditioning regimens were used in 18 (43%) and 24 (57%) patients, respectively. Transplant source was bone marrow (n = 30, 71%) and peripheral blood (n = 12, 29%).
Diagnosis and classification of cGHVD was based on established NIH criteria [16]. Mild, moderate and severe cGVHD was observed in three (7%), 15 (36%) and 24 (57%) patients, respectively. Classical cGVHD was diagnosed in 36 (86%) children and overlap syndrome in six (14%). Among children with mild cGVHD, one patient received therapy due to non-malignant disease and awareness of cGVHD progression. In two other cases, systemic immunosuppressive therapy (IST) was initiated by decision of attending physician. Several organs were affected in 32 (76%) patients and only one organ, in 10 cases (24%). In a majority of patients, skin (n = 32, 76%), mucous membranes (n = 22, 52%), liver (n = 10, 24%) and lungs (n = 9, 21%) were involved. Less common presentations included gut (n = 5, 12%) and joints (n = 3, 7%).
First line therapy of cGVHD was methylprednisolone 1 mg/kg/day in all patients with further gradual tapering according to response and tolerance. Criteria for SR cGVHD were progression despite this approach for 1–2 weeks, stable disease while on 0.5 ≥ mg/kg/day for 1 month or inability to withdraw steroids without flare of the disease. Four patients (10%) had intolerance to steroids due to unacceptable toxicity (infectious or metabolic complications). SR cGVHD was diagnosed in 38 children (90%). ECP was administered in patients on individual basis after discussion on board of doctors.
All the patients received ECP as second (n = 18, 43%) or third (n = 24, 57%) line of therapy. Initial ECP therapy consisted of bimonthly treatment schedule for two consecutive days with an opportunity of further tapering, according to clinical response. Any concurrent treatment administered before ECP could be continued if considered necessary. ECP was co-administered with ruxolitinib in eight (19%) children, sirolimus in five (12%), imatinib in six (14%), rituximab in two (5%) and ibrutinib in one (2%).Detailed characteristic of concomitant IST and requirement for systemic therapy escaltion after ECP is shown in Table 1. All the patients received off-line ECP procedures with a MacoGenic device (Macopharma, Lille, France) according to the standard protocol [31]. Apheresis was performed with a Cobe Spectra device (Terumo, Lakewood, CA, USA). Sodium citrate was the anticoagulant used during mononuclear cell collection. The final volume of the product was 150 mL. It was further diluted with normal saline up to 300 mL. Afterwards a photosensitizing agent (8-methoxypsoralen) was added to the product up to concentration of 200 ng/mL with subsequent ultraviolet irradiation (320–400 nm) of 2 J/cm2. Total duration of the procedure was approximately 3 h.Median number of ECP procedures was 10 (2–46), with total number of 606, and median duration for 4 months (1–42). Median interval from allo-HSCT to first ECP was 7 months (4–41). Clinical response to ECP-based therapy was evaluated by means of established criteria [32]. Complete response was diagnosed in patients with resolution of all cGVHD symptoms. Partial response (PR) was diagnosed in case of improvement in ≥1 organ or site without any signs of deterioration in other localizations. PR was diagnosed after decrease in clinician overall severity score by ≥2 points according to the 2014 Response Criteria Working Group Report. Progression of cGVHD was defined separately for each organ based on the above mentioned recommendations. Patients that did not meet criteria of CR, PR or progression were classified as unchanged. If response to ECP-based therapy was registered than patients were gradually weaned away from steroids at a rate of 25% dose reduction biweekly. Simultaneously with steroid tapering the ECP schedule was reevaluated every 3 months with the switch to monthly or quarterly procedures in responders on an individual basis. Overall survival (OS), progression-free survival (PFS) and failure-free survival (FFS) were calculated by Kaplan-Meier method. Failure-free survival was defined as the absence of relapse, non-relapse mortality or addition of another systemic therapy. Events for FFS were considered relapse, death, or IST escalation. Non-relapse mortality (NRM) and cumulative incidence of relapse (CIR) were assessed using competing risks. Univariate analysis was performed by Cox regression. The Mann-Whitney U test was used to compare whether there was difference in the dependent variable for two independent groups. Variables related to patient, disease, and transplants were characterized using descriptive statistics. For statistical analysis we used the Easy R (EZR) program [33].

5. Conclusions

ECP remains an important treatment option in children with SR cGVHD, despite emergence of new effective options, due to safety and high ORR, with similar clinical efficiency in pediatric and adult patients. To our knowledge this is probably the first publication demonstrating FFS in children with cGVHD after ECP. Suboptimal FFS represents potential of ECP-based therapy to cure only third of children with cGVHD. A significant proportion of patients would require escalation of IST to control cGVHD. This means that ECP should not be regarded as a game changer but as an important approach that can help to relieve symptoms in most of the patients at least transiently.

Author Contributions

Conceptualization, A.K. (Andrey Kozlov) and L.Z.; methodology, O.P.; resources, methodology M.E.; software, T.B. and A.O.; formal analysis, P.K.; investigation, Z.R. and I.S.; resources, E.M. and M.E.; data curation, A.A. and I.K.; writing—original draft preparation, A.K. (Andrey Kozlov), A.G. and A.D.; writing—review and editing, I.M. and A.C.; supervision, A.K. (Alexander Kulagin); project administration, S.B. and E.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Ethics Committee of RM Gorbacheva Research Institute, Pavlov University (St. Petersburg, Russia).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data are not publicly available due to privacy and ethical restrictions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Inagaki, J.; Moritake, H.; Nishikawa, T.; Hyakuna, N.; Okada, M.; Suenobu, S.-I.; Nagai, K.; Honda, Y.; Shimomura, M.; Fukano, R.; et al. Long-Term Morbidity and Mortality in Children with Chronic Graft-versus-Host Disease Classified by National Institutes of Health Consensus Criteria after Allogeneic Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2015, 21, 1973–1980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Luznik, L.; O’Donnell, P.V.; Fuchs, E.J. Post-Transplantation Cyclophosphamide for Tolerance Induction in HLA-Haploidentical Bone Marrow Transplantation. Semin. Oncol. 2012, 39, 683–693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Pirogova, O.V.; Moiseev, I.S.; Alyansky, A.L.; Babenko, E.V.; Darskaya, E.I.; Slesarchuk, O.A.; Bondarenko, S.N.; Afanasyev, B.V. Risk-adapted graft-versus host disease prophylaxis with post-transplantation cyclophosphamide in related, unrelated and haploidentical stem cell transplantations. Cell. Ther. Transplant. 2016, 5, 54–56. [Google Scholar] [CrossRef] [Green Version]
  4. Modi, B.; Hernandez-Henderson, M.; Yang, D.; Klein, J.; Dadwal, S.; Kopp, E.; Huelsman, K.; Mokhtari, S.; Ali, H.; Al Malki, M.M.; et al. Ruxolitinib as Salvage Therapy for Chronic Graft-versus-Host Disease. Biol. Blood Marrow Transplant. 2019, 25, 265–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Jagasia, M.; Perales, M.-A.; Schroeder, M.A.; Ali, H.; Shah, N.N.; Chen, Y.-B.; Fazal, S.; Dawkins, F.W.; Arbushites, M.C.; Tian, C.; et al. Ruxolitinib for the treatment of steroid-refractory acute GVHD (REACH1): A multicenter, open-label phase 2 trial. Blood 2020, 135, 1739–1749. [Google Scholar] [CrossRef] [PubMed]
  6. Ferrara, J.L.; Levine, J.; Reddy, P.; Holler, E. Graft-versus-host disease. Lancet 2009, 373, 1550–1561. [Google Scholar] [CrossRef]
  7. Kolb, H.-J. Graft versus host disease: From experiments to clinical insight. Cell. Ther. Transplant. 2010, 2, 3. [Google Scholar]
  8. Cooke, K.R.; Luznik, L.; Sarantopoulos, S.; Hakim, F.T.; Jagasia, M.; Fowler, D.H.; Brink, M.R.V.D.; Hansen, J.A.; Parkman, R.; Miklos, D.B.; et al. The Biology of Chronic Graft-versus-Host Disease: A Task Force Report from the National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease. Biol. Blood Marrow Transplant. 2017, 23, 211–234. [Google Scholar] [CrossRef] [Green Version]
  9. Heshmati, F. Updating ECP action mechanisms. Transfus. Apher. Sci. 2014, 50, 330–339. [Google Scholar] [CrossRef]
  10. Besnier, D.P.; Chabannes, D.; Mahe, B.; Mussini, J.M. Treatment of graft-versus-host disease by extracorpo-real photochemotherapy: A pilot study. Transplantation 1997, 64, 49–54. [Google Scholar] [CrossRef]
  11. Knobler, R.; Arenberger, P.; Arun, A.; Assaf, C.; Bagot, M.; Berlin, G.; Bohbot, A.; Calzavara-Pinton, P.; Child, F.; Cho, A.; et al. European dermatology forum—Updated guidelines on the use of extracorporeal photopheresis 2020—Part 1. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 2693–2716. [Google Scholar] [CrossRef]
  12. Flowers, M.E.D.; Apperley, J.F.; van Besien, K.; Elmaagacli, A.; Grigg, A.; Reddy, V.; Bacigalupo, A.; Kolb, H.-J.; Bouzas, L.; Michallet, M.; et al. A multicenter prospective phase 2 randomized study of extracorporeal photopheresis for treatment of chronic graft-versus-host disease. Blood 2008, 112, 2667–2674. [Google Scholar] [CrossRef]
  13. Jagasia, M.; Scheid, C.; Socié, G.; Ayuk, F.A.; Tischer, J.; Donato, M.L.; Bátai, Á.; Chen, H.; Chen, S.-C.; Chin, T.; et al. Randomized controlled study of ECP with methoxsalen as first-line treatment of patients with moderate to severe cGVHD. Blood Adv. 2019, 3, 2218–2229. [Google Scholar] [CrossRef] [Green Version]
  14. McKenna, K.E.; Whittaker, S.; Rhodes, L.; Taylor, P.; Lloyd, J.; Ibbotson, S.; Russell-Jones, R. Evidence-based practice of photopheresis 1987–2001: A report of a workshop of the British Photodermatology Group and the U.K. Skin Lymphoma Group. Br. J. Dermatol. 2005, 154, 7–20. [Google Scholar] [CrossRef]
  15. Sarantopoulos, S.; Cardones, A.R.; Sullivan, K.M. How I treat refractory chronic graft-versus-host disease. Blood 2019, 133, 1191–1200. [Google Scholar] [CrossRef] [Green Version]
  16. Jagasia, M.H.; Greinix, H.T.; Arora, M.; Williams, K.M.; Wolff, D.; Cowen, E.W.; Palmer, J.; Weisdorf, D.; Treister, N.S.; Cheng, G.-S.; et al. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group Report. Biol. Blood Marrow Transplant. 2015, 21, 389–401.e1. [Google Scholar] [CrossRef] [Green Version]
  17. Waller, E.K.; Miklos, D.; Cutler, C.; Arora, M.; Jagasia, M.H.; Pusic, I.; Flowers, M.E.; Logan, A.C.; Nakamura, R.; Chang, S.; et al. Ibrutinib for Chronic Graft-versus-Host Disease After Failure of Prior Therapy: 1-Year Update of a Phase 1b/2 Study. Biol. Blood Marrow Transplant. 2019, 25, 2002–2007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Maas-Bauer, K.; Kiote-Schmidt, C.; Bertz, H.; Apostolova, P.; Wäsch, R.; Ihorst, G.; Finke, J.; Zeiser, R. Ruxolitinib–ECP combination treatment for refractory severe chronic graft-versus-host disease. Bone Marrow Transplant. 2021, 56, 909–916. [Google Scholar] [CrossRef] [PubMed]
  19. Greinix, H.T.; Worel, N.; Just, U.; Knobler, R. Extracorporeal photopheresis in acute and chronic graft-versus-host disease. Transfus. Apher. Sci. 2014, 50, 349–357. [Google Scholar] [CrossRef]
  20. Jacobsohn, D.A.; Arora, M.; John, P.K.; Hassebroek, A.; Flowers, M.E.; Cutler, C.S.; Ur-bano-Ispizua, A.; Bolwell, B.J.; Antin, J.H.; Boyiadzis, M.; et al. Risk factors associated with increased nonrelapse mortality and with poor overall sur-vival in children with chronic graft-versus-host disease. Blood 2011, 118, 4472–4479. [Google Scholar] [CrossRef]
  21. Perotti, C.; Del Fante, C.; Tinelli, C.; Viarengo, G.; Scudeller, L.; Zecca, M.; Locatelli, F.; Salvaneschi, L. Extracorporeal photochemotherapy in graft-versus-host disease: A longitudinal study on factors influencing the response and survival in pediatric patients. Transfusion 2010, 50, 1359–1369. [Google Scholar] [CrossRef] [PubMed]
  22. Lucchini, G.; Labopin, M.; Beohou, E.; Dalissier, A.; Dalle, J.H.; Cornish, J.; Zecca, M.; Samarasinghe, S.; Gibson, B.; Locatelli, F.; et al. Impact of Conditioning Regimen on Outcomes for Children with Acute Myeloid Leukemia Undergoing Transplantation in First Complete Remission. An Analysis on Behalf of the Pediatric Disease Working Party of the European Group for Blood and Marrow Transplantation. Biol. Blood Marrow Transplant. 2017, 23, 467–474. [Google Scholar] [CrossRef] [Green Version]
  23. Paina, O.V.; Kozhokar, P.V.; Borovkova, A.S.; Frolova, A.S.; Ekushov, K.A.; Bykova, T.A.; Rakhmanova, Z.Z.; Galas, M.A.; Khabirova, A.G.; Markova, I.V.; et al. Ten year expierence of allogeneic haploaidentical hematopoietic stem cell transplantation with non-manipulated grafts in children and adolescents with high risk acute leukemia. Cell. Ther. Transplant. 2018, 7, 20–27. [Google Scholar] [CrossRef] [Green Version]
  24. Beynarovich, A.V.; Babenko, E.V.; Moiseev, I.S.; Paina, O.V.; Pirogova, O.V.; Rudakova, T.A.; Gindina, T.L.; Darskaya, E.I.; Morozova, E.V.; Bondarenko, S.N.; et al. Haploidentical stem cell transplantation in adults for the treatement of hematological diseases: Results of a single center (CIC 725). Cell. Ther. Transplant. 2019, 8, 26–35. [Google Scholar] [CrossRef]
  25. Apisarnthanarax, N.; Donato, M.L.; Korbling, M.; Couriel, D.R.; Gajewski, J.L.; Giralt, S.; Khouri, I.F.; Hosing, C.; E Champlin, R.; Duvic, M.; et al. Extracorporeal photopheresis therapy in the management of steroid-refractory or steroid-dependent cutaneous chronic graft-versus-host disease after allogeneic stem cell transplantation: Feasibility and results. Bone Marrow Transplant. 2003, 31, 459–465. [Google Scholar] [CrossRef] [Green Version]
  26. Flinn, A.M.; Macheka, S.; Slatter, M.; Ewins, A.; Gibson, B.; Lawson, S.; Tailby, A.; Lucchini, G.; New, H.; James, B.; et al. A survey of extracorporeal photopheresis treatment in pediatric patients in the United Kingdom. eJHaem 2020, 1, 293–296. [Google Scholar] [CrossRef]
  27. Buense, R.; Duarte, I.A.G.; Bouer, M. Localized scleroderma: Assessment of the therapeutic response to phototherapy. An. Bras. Dermatol. 2012, 87, 63–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. DeSimone, R.A.; Schwartz, J.; Schneiderman, J. Extracorporeal photopheresis in pediatric patients: Practical and technical considerations. J. Clin. Apher. 2017, 32, 543–552. [Google Scholar] [CrossRef] [PubMed]
  29. Schreiner, T.; Gaczkowski, A.; Scharffetter-Kochanek, K.; Borberg, H. Small-scale extracorporeal photopheresis for the treatment of cutaneous T-cell lymphoma: A report of 3 cases. Transfus. Apher. Sci. 2005, 32, 197–203. [Google Scholar] [CrossRef]
  30. Messina, C.; Locatelli, F.; Lanino, E.; Uderzo, C.; Zacchello, G.; Cesaro, S.; Pillon, M.; Perotti, C.; Del Fante, C.; Faraci, M.; et al. Extracorporeal photochemotherapy for paediatric patients with graft-versus-host disease after haematopoietic stem cell transplantation. Br. J. Haematol. 2003, 122, 118–127. [Google Scholar] [CrossRef]
  31. Heshmati, F Mechanisms of action of extracorporeal photochemotherapy. Transfus. Apher. Sci. 2003, 29, 61–70. [CrossRef]
  32. Lee, S.J.; Wolff, D.; Kitko, C.; Koreth, J.; Inamoto, Y.; Jagasia, M.; Pidala, J.; Olivieri, A.; Martin, P.J.; Przepiorka, D.; 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. The 2014 Response Criteria Working Group Report. Biol. Blood Marrow Transplant. 2015, 21, 984–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kanda, Y. Investigation of the freely available easy-to-use software ‘EZR’ for medical statistics. Bone Marrow Transplant. 2013, 48, 452–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Progression-free survival (1) and overall survival (2) in children with cGVHD after ECP.
Figure 1. Progression-free survival (1) and overall survival (2) in children with cGVHD after ECP.
Pharmaceuticals 14 00808 g001
Figure 2. Non-relapse mortality (1) and cumulative incidence of relapse (2) in children with cGVHD after ECP.
Figure 2. Non-relapse mortality (1) and cumulative incidence of relapse (2) in children with cGVHD after ECP.
Pharmaceuticals 14 00808 g002
Figure 3. Failure-free survival in children with cGVHD after ECP.
Figure 3. Failure-free survival in children with cGVHD after ECP.
Pharmaceuticals 14 00808 g003
Table 1. Demographics of patients with cGVHD treated with ECP.
Table 1. Demographics of patients with cGVHD treated with ECP.
AgeDiagnosiscGVHD SeverityAffected OrgansSteroids Prior ECP, mg/kg/dayNumber of ECP ProceduresResponse to ECPSteroids Post ECP, mg/kg/dayConcomitant ISTRequirement for IST Escalation after ECPStatus
(Cause)
Follow-Up,
Days
113MDSsevereskin, mucous membranes0.58CR0.1nonoalive1466
25AMLmoderateskin0.45CR0CsAnoalive4541
316ALLmoderateskin, lungs012PR0CsAnoalive4064
410ALLsevereskin17progression1Txyesdead (ALL)180
510ALLmoderateskin0.54progression2ruxolitinibyesalive721
618ALLsevereskin, mucous membranes0.34PR0.3ibrutinibnoalive463
710BALmoderateskin, mucous membranes, lungs 0.58PR0.2nonodead (ALL)1097
813ALLmoderatejoints, gut0.66PR0.25CsAnoalive774
95AMLmoderatemucous membranes0.5 PR0.5nonoalive256
1013BALsevereliver0.310PR0sirolimusnodead (BAL)396
118CMLseveremucous membranes124PR0.25imatinibnoalive3929
1212AMLsevereskin, liver, lungs0.720CR0.1imatinibyesalive1834
131ALLsevereskin, lungs0.252PR0.1CsAnoalive3394
1413ALLmoderateskin, liver0.42PR0.4ruxolitinibnodead (ALL)352
153Hurlermildskin, mucous membranes0.56PR0.2ruxolitinibyesalive864
165AMLmoderateskin, gut0.526PR0.3imatinibnoalive1217
1716AMLsevereskin, mucous membranes, liver, lungs 0.433unchanged0.75rituximabyesdead (cGVHD)604
1818AMLseveremucous membranes, liver 0.612PR0.25Txnoalive3683
1913ALLmildskin, mucous membranes 15PR0.5sirolimusyesdead (cGVHD)201
2016AMLmildskin, liver 0.828PR0Txnoalive1379
214Fanconisevereskin, gut0.56PR0.25Txnoalive3140
2211ALLsevereskin0.424CR0.1ruxolitinibnoalive1334
2314Fanconisevereskin, joints, lungs 0.612PR0.4ruxolitinibnodead (Fanconi)259
241AMLsevereskin, mucous membranes, liver0.2510PR0.1ruxolitinibnoalive1453
258ALLmoderateskin, gut, liver 0.718PR0.25Tx, mmfnodead (ALL)182
2615AMLsevereskin, mucous membranes, lungs 0.42progression0.5Txyesdead (cGVHD)249
277ALLmoderatemucous membranes0.637PR0.8sirolimusyesalive648
286ALLsevereskin, mucous membranes, lungs 035unchanged0rituximabyesdead (cGVHD)587
296ALLsevereskin, mucous membranes, gut, liver 040PR0imatinibyesalive1246
303BALmoderateskin, mucous membranes0.44CR0Txnoalive4167
3117BALseveremucous membranes 0.310progression0imatinibyesdead (BAL)325
3213ALLmoderatemucous membranes0.512PR0.25sirolimusnoalive 342
3315ALLmoderateskin134CR0.1 noalive400
347JMMLmoderateskin0.255CR0.1Txnoalive2800
3516HLseveremucous membranes0.414PR0.25ruxolitinibyesalive590
3617ALLseveremucous membranes0.75progression0.7CsAyesdead (ALL)252
3716CMLsevereskin, joints0.334PR0.3CsAyesalive3568
3817HLsevereskin, mucous membranes, lungs 0.546unchanged0.5ruxolitinibyesalive536
399JMLLmoderateskin, mucous membranes0.442PR0.25sirolimusnoalive2146
4016AMLsevereskin, mucous membranes 010progression0imatinib, Txyesdead (AML)870
4118ALLsevereskin0.48progression0Tx, mmfyesdead (ALL)270
4217AMLsevereskin, liver0.58progression0.75noyesdead (cGVHD)239
MDS—myelodysplastic syndrome, AML—acute myeloid leukemia, ALL—acute lymphoblastic leukemia, BAL—biphenotypic acute leukemia, JMML—juvenile meylomonocytic leukemia, HL—Hodgkin lymphoma, CML—chronic myelogenous leukemia, ECP—extracorporeal photopheresis, CR—complete response, PR—partial response, IST—immunosupressive therapy, CsA—cyclosporine A, Tx—tacrolimus, mmf—mycophenolate mofetil, cGVHD—chronic graft versus host disease.
Table 2. Clinical response to extracorporeal photophersis in children with cGVHD.
Table 2. Clinical response to extracorporeal photophersis in children with cGVHD.
Efficiency of ECP in Children with cGVHD
ResponseSkin
(n = 32)
Mucous Membrane
(n = 22)
Lungs
(n = 9)
Liver
(n = 10)
Gut
(n = 5)
Joints
(n = 3)
Overall Response
(n = 42)
Complete response, n (%)7 (22%)2 (9%)1 (11%)2 (20%)N/AN/A7 (17%)
Partial response, n (%)17 (53%)14 (64%)1 (11%)6 (60%)4 (80%)2 (67%)24 (57%)
Unchanged, n (%)4 (12.5%)3 (13.5%)6 (67%)1 (10%)1 (20%)1 (33%)3 (7%)
Progression, n (%)4 (12.5%)3 (13.5%)1 (11%)1 (10%)N/AN/A8 (19%)
N/A—not applicable.
Table 3. An univariate analysis of factors influencing survival.
Table 3. An univariate analysis of factors influencing survival.
VariableHR95%CIp
Response to ECP 7.82.7–23.10.0002
Sex2.30.6–8.70.2
Disease risk group (HR vs. SR)1.9649–0.40.4
Time interval from HSCT to ECP0.40.1–1.50.2
Ruxolitinib− vs. Ruxolitinib+1.70.5–6.90.4
ECP monotherapy vs. ECP in combination therapy0.60.2–2.40.5
cGVHD severity (severe vs. moderate)2.6560.8–8.30.1
ECP—extracorporeal photopheresis, HR—high risk, SR—standard risk, HSCT—hematopoietic stem cell transplantation, cGVHD—chronic graft versus host disease, ruxolitinib− (absence of ruxolitinib in combination therapy), ruxolitinib+ (presence of ruxolitinib in combination therapy).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kozlov, A.; Estrina, M.; Paina, O.; Bykova, T.; Osipova, A.; Kozhokar, P.; Rakhmanova, Z.; Solodova, I.; Morozova, E.; Alyansky, A.; et al. Extracorporeal Photopheresis in Children with Chronic Graft-Versus-Host Disease. Pharmaceuticals 2021, 14, 808. https://doi.org/10.3390/ph14080808

AMA Style

Kozlov A, Estrina M, Paina O, Bykova T, Osipova A, Kozhokar P, Rakhmanova Z, Solodova I, Morozova E, Alyansky A, et al. Extracorporeal Photopheresis in Children with Chronic Graft-Versus-Host Disease. Pharmaceuticals. 2021; 14(8):808. https://doi.org/10.3390/ph14080808

Chicago/Turabian Style

Kozlov, Andrey, Maria Estrina, Olesia Paina, Tatiana Bykova, Anna Osipova, Polina Kozhokar, Zhemal Rakhmanova, Irina Solodova, Elena Morozova, Alexander Alyansky, and et al. 2021. "Extracorporeal Photopheresis in Children with Chronic Graft-Versus-Host Disease" Pharmaceuticals 14, no. 8: 808. https://doi.org/10.3390/ph14080808

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop