ArticlePDF Available

Daratumumab for Treatment of Antibody-Mediated Rejection after ABO-Incompatible Kidney Transplantation

Authors:

Abstract and Figures

We report the effectiveness of daratumumab, a human IgGκ monoclonal antibody targeting CD38 on plasma cells, for therapy-refractory antibody-mediated rejection (AMR) due to blood group antibodies in a 59-year-old man who received a living ABO-incompatible kidney transplantation. Standard treatment options for AMR due to blood group antibodies including immunoadsorption, lymphocyte depletion with anti-human T-lymphocyte globulins, intravenous methylprednisolone pulses and eculizumab limited tissue injury, however failed to sufficiently suppress blood group antibody production. After administration of daratumumab as a rescue therapy, blood group antibody titers decreased and remained at low levels without further immunoadsorption and allowed kidney graft function to recover.
Content may be subject to copyright.
Case Rep Nephrol Dial
DOI: 10.1159/000503951
© 2019 The Author(s)
Published by S. Karger AG, Basel
www.karger.com/cnd
This article is licensed under the Creative Commons Attribution-NonCommercial 4.0
International License (CC BY-NC) (http://www.karger.com/Services/OpenAccessLicense).
Usage and distribution for commercial purposes requires written permission.
Patricia Hirt-Minkowski, MD
Clinic for Transplantation Immunology and Nephrology, University Hospital Basel
Petersgraben 4
CH4031 Basel (Switzerland)
E-Mail patricia.hirt-minkowski@usb.ch
Case Report
Daratumumab for Treatment of
Antibody-Mediated Rejection after
ABO-Incompatible Kidney
Transplantation
Davide Spicaa Till Junkerb Michael Dickenmanna Stefan Schauba, c, d
Jürg Steigera, c Tanja Rüflie Jörg Halterb Helmut Hopferf
Andreas Holbrob, e Patricia Hirt-Minkowskia
aTransplantation Immunology and Nephrology, University Hospital Basel,
Basel, Switzerland; bDivision of Hematology, Department of Medicine, University Hospital
Basel, Basel, Switzerland; cTransplantation Immunology, Department of Biomedicine,
University of Basel, Basel, Switzerland; dHLA-Diagnostic and Immunogenetics, Department
of Laboratory Medicine, University Hospital Basel, Basel, Switzerland; eBlood Transfusion
Service, Swiss Red Cross, Basel, Switzerland; fInstitute for Pathology, University Hospital
Basel, Basel, Switzerland
Keywords
Antibody-mediated rejection · ABO-incompatible kidney transplantation · Anti-rejection
therapy · Daratumumab
Abstract
We report the effectiveness of daratumumab, a human IgGκ monoclonal antibody targeting
CD38 on plasma cells, for therapy-refractory antibody-mediated rejection (AMR) due to blood
group antibodies in a 59-year-old man who received a living ABO-incompatible kidney trans-
plantation. Standard treatment options for AMR due to blood group antibodies including im-
munoadsorption, lymphocyte depletion with anti-human T-lymphocyte globulins, intravenous
methylprednisolone pulses and eculizumab limited tissue injury, however failed to sufficiently
suppress blood group antibody production. After administration of daratumumab as a rescue
Case Rep Nephrol Dial
DOI: 10.1159/000503951
© 2019 The Author(s). Published by S. Karger AG, Basel
www.karger.com/cnd
Spica et al.: Daratumumab for Treatment of Antibody-Mediated Rejection after ABO-
Incompatible Kidney Transplantation
138
therapy, blood group antibody titers decreased and remained at low levels without further
immunoadsorption and allowed kidney graft function to recover.
© 2019 The Author(s)
Published by S. Karger AG, Basel
Introduction
Kidney transplantation is the therapy of choice for many patients with end-stage renal
disease. Pretransplant risk stratification and modern immunosuppression have led to striking
advances over the past decades, resulting in better long-term graft survival [14]. Neverthe-
less, there still exist important unmet clinical needs and problems to be solved for kidney
transplant recipients [57]. Antibody-mediated rejection (AMR) has been recognized as one
of the most important causes of graft loss [810]. Although less frequent than antibodies di-
rected against human leukocyte antigens (i.e., donor specific HLA-antibodies [DSA]) [11], an-
tibodies against ABO blood group antigens can be the cause of AMR [12]. In a meta-analysis,
which included 26 centers, describing 1,346 ABO-incompatible kidney transplant patients,
AMR within the first year post-transplant was more common and occurred with a fourfold
risk compared to 4,943 center-matched controls, who were ABO-compatible [12]. To date,
there are no US Food and Drug Administration (FDA)-approved immunosuppressive drugs
for either prevention or treatment of AMR [13]. The only FDA-approved drugs for treatment
of acute rejection are polyclonal anti-T lymphocyte globulins [6]. Several additional agents
have been employed as off-label for the treatment of AMR with variable effectiveness such as
intravenous immunoglobulins, rituximab, bortezomib, and eculizumab as well as removing of
circulating anti-HLA antibodies and blood group antibodies by plasmapheresis/plasma ex-
change or immunoadsorption [13, 14]. Thus, our armamentarium to treat AMR is still incom-
plete. For this reason, new therapeutic options to reduce the burden of AMR are urgently
needed.
Daratumumab is a human monoclonal IgGκ antibody targeting the glycoprotein CD38
which is expressed on plasma cells and induces cell death and apoptosis through different
immune effector mechanisms such as complement-mediated cytotoxicity, antibody-mediated
cytotoxicity, antibody-dependent phagocytosis and direct induction of apoptosis [15]. Based
on data of the MMY2002 (SIRIUS) [16] and the GEN501 [17] studies, daratumumab has
emerged as a new option in therapy-refractory multiple myeloma and was approved for treat-
ment of multiple myeloma in 2015 [18]. As CD38 is also expressed on nonmalignant plasma
cells, it seems obvious to test daratumumab as a rescue therapy in other diseases with patho-
genetically involved, antibody-producing plasma cells after failure of established therapies.
Thus, daratumumab has been used in other settings where antibody production by plasma
cells is the pathophysiologic key feature. The effective treatment of refractory post-transplant
autoimmune hemolytic anemia after allogenic hematopoietic stem cell transplantation
(HSCT) was reported by Schuetz et al. [19] in a small case series of 3 patients (1 adult, 2 pedi-
atric cases). Furthermore, daratumumab was used successfully for treatment of delayed red-
cell engraftment (i.e., pure red cell aplasia) due to ABO incompatibility after allogenic HSCT
[20]. These conditions share the central role of deleterious blood group antibodies produced
by nonmalignant plasma cells that target and destroy allogenic tissue. In all reported cases,
depletion of the plasma cells by daratumumab resulted in resolution of red blood cell destruc-
tion [19, 20].
Case Rep Nephrol Dial
DOI: 10.1159/000503951
© 2019 The Author(s). Published by S. Karger AG, Basel
www.karger.com/cnd
Spica et al.: Daratumumab for Treatment of Antibody-Mediated Rejection after ABO-
Incompatible Kidney Transplantation
139
As therapeutic options for AMR are limited, an anti-CD38 agent such as daratumumab
may be a new treatment option to be evaluated in patients with no response to so far utilized
anti-rejection therapies for AMR.
Case Report
A 59-year-old man with end-stage renal disease due to IgA nephropathy received a living
kidney donation from his 64-year-old HLA-haploidentical sister. HLA antibody screening be-
fore transplantation performed by Luminex single HLA antigen beads (LabScreen SA; One
Lambda Inc., CA, USA) showed no DSA. Because of major ABO incompatibility (i.e., recipient
with blood group B and donor with blood group A), the recipient was treated with rituximab
375 mg/m2 for B-cell depletion 4 weeks before transplantation. Baseline immunosuppressive
therapy with tacrolimus (tac) with a goal to reach a trough level of 8−10 µg/L and mycophe-
nolate mofetil (MMF) twice daily 500 mg was started 2 weeks before transplantation and ex-
tended by prednisone with an initial dose of 30 mg per day on day −6 (i.e., 6 days before trans-
plantation). During the in-hospital stay, tac trough levels were measured three times per week
(i.e., within the first 37 days post-transplant), followed by weekly measurements thereafter.
In addition, from day −5 to −1 (i.e., from 5 days to 1 day before transplantation) the patient
received five sessions of immunoadsorption with a Glycosorb® column (Glycorex Transplan-
tation AB, Lund, Sweden) to remove circulating anti-A blood group antibodies. When starting
immunoadsorption, anti-A IgM isohemagglutinin and anti-A IgG titers were 1:8 and 1:32, re-
spectively. On the day of transplantation, anti-A IgM isohemagglutinin and anti-A IgG titers
had been reduced to 1:2 and 1:4, respectively (Fig. 1). Induction immunosuppression con-
sisted of basiliximab (i.e., 20 mg at day 0, followed by 20 mg at day 4) and baseline immuno-
suppressive therapy with tac, MMF, and prednisone was continued during the whole course.
This treatment approach reflects the standard procedure in ABO-incompatible kidney trans-
plantation at our center as described before [21]. Serum creatinine levels dropped from 604
to 189 µmol/L at day 4 (Fig. 1), reaching an adequate baseline estimated GFR (eGFR) of 33
mL/min/1.73 m2 calculated by CKD-EPI formula, taking into account the patient’s/donor’s
age, weight mismatch, and kidney function of the sister before donation (i.e., eGFR of 94
mL/min/1.73 m2).
In the first days after transplantation, anti-A IgM and anti-A IgG isohemagglutinin titers
remained low (maximal 1:2 and 1:4, respectively). On day 6, both titers began to rise and
reached a maximum of 1:64, which was followed by a sudden drop in kidney graft function
(decrease in eGFR from 33 to 14 mL/min/1.73 m2 on day 10) (Fig. 1). A biopsy on day 7
showed mild diffuse acute tubular damage (Fig. 2a). By immunofluorescence, both IgM and
C3c deposition was seen in the peritubular capillaries in addition to the C4d positivity usually
seen in AB0-incompatible graft biopsies suggesting very recent antibody-mediated comple-
ment activation (Fig. 2bd). In the context of rising anti-A isohemagglutinins and declining
graft function, these findings were interpreted as a very early episode of AMR due to anti-A
blood group antibodies. Immunoadsorption was restarted on day 8, effectively removing the
circulating anti-A blood group antibodies (Fig. 1). To suppress further antibody production,
anti-human T-lymphocyte globulins (i.e., totally seven infusions with a dosage between 2 and
4 mg/kg of body weight on day 9, from day 14 to 18, and on day 20) as well as high-dose
intravenous methylprednisolone (i.e., totally five pulses between day 7 and 11) were admin-
istrated. Afterwards, prednisone dosed 0.5 mg/kg body weight was continued. Additionally,
eculizumab, a monoclonal antibody that targets complement C5 (i.e., three infusions of 900
Case Rep Nephrol Dial
DOI: 10.1159/000503951
© 2019 The Author(s). Published by S. Karger AG, Basel
www.karger.com/cnd
Spica et al.: Daratumumab for Treatment of Antibody-Mediated Rejection after ABO-
Incompatible Kidney Transplantation
140
mg per week on days 12, 19 and 26, respectively), was administrated to limit destruction of
endothelial cells by complement-dependent cytotoxicity. So far, our approach for treatment
of AMR stabilized kidney graft function with low eGFR levels between 18 and 20 mL/min/1.73
m2. Unfortunately, anti-A antibody titers immediately rose again after discontinuation of im-
munoadsorption on day 19 to IgM isohemagglutinin titers of 1:32 and IgG of 1:32 on day 23
post-transplant. Thus, immunoadsorption of anti-A blood group antibodies was resumed and
anti-A antibodies could successfully be removed. When immunoadsorption was stopped once
again on day 26, anti-A antibody titers rose repeatedly, reaching IgM isohemagglutinin titers
of 1:16 and IgG of 1:16, respectively on day 29, as summarized in Figure 1.
To prevent long-term dependence of immunoadsorption and eventually graft failure and
due to the lack of effectiveness of the so far established combined therapy, daratumumab was
administrated as a rescue therapy at a dose of 16 mg per kg of body weight in weekly intervals
beginning on day 30 after kidney transplantation. The patient gave his consent after infor-
mation about the experimental approach. Eculizumab was discontinued avoiding interference
with daratumumab by possibly blocking complement-mediated cytotoxicity of the targeted
plasma cells, which is considered an important mechanism of action of daratumumab. Within
72 h after the first dose of daratumumab, the pathogenic IgM isohemagglutinins started to fall
continuously without any further immunoadsorption. In total, the patient received six infu-
sions of daratumumab, without any severe drug-related adverse event. Two weeks after ad-
ministration of the last daratumumab infusion on day 65, the anti-A IgM isohemagglutinin and
anti-A IgG titers were 1:1 and 1:2, respectively and persisted at low levels the following weeks.
As binding of daratumumab to CD38 on red blood cells can affect measurement of anti-A IgG
antibodies, anti-A IgG titers were validated after incubation with dithiothreitol as described
previously [22]. A biopsy of the kidney graft on day 73 post-transplant showed no signs of
acute rejection (image not shown). The patient’s graft function nearly recovered to baseline
with eGFR levels at 2732 mL/min/1.73 m2. To note, no further immunoadsorption was nec-
essary after initiation of daratumumab.
Discussion
To the best of our knowledge, this is the first report of successful administration of dara-
tumumab for treatment of therapy-refractory AMR in kidney transplantation. In the context
of rising anti-A isohemagglutinin titers right before graft function deterioration, early active
ABO blood group AMR was strongly suggested. In this case, it was confirmed by the histologic
findings of tissue injury (unexplained acute tubular injury) and evidence of interaction of IgM
antibodies with vascular endothelial cells on the peritubular capillaries with concomitant ac-
tivation and deposition of complement factor C3c. This provided the pathogenetic linkage be-
tween the rising anti-A isohemagglutinins and kidney transplant injury. Thus, we interpreted
the early rise in anti-A isohemagglutinin titers as clinically relevant. Because of the evidence
for early anti-A isohemagglutinins-mediated tissue injury, we did not screen for de novo DSA
in this patient, who was tested negative for pre-transplant DSA timely before transplantation
and treated with rituximab for desensitization 4 weeks prior to transplantation.
In summary, our patient developed early active AMR caused by anti-A IgM isohemagglu-
tinins after kidney transplantation from his ABO-incompatible sister despite following estab-
lished therapeutic strategies for ABO-incompatible kidney transplantation [21], and immedi-
ate treatment with anti-human T-lymphocyte globulins, high-dose methylprednisolone,
blocking the complement-mediated effector mechanisms of tissue injury by eculizumab and
Case Rep Nephrol Dial
DOI: 10.1159/000503951
© 2019 The Author(s). Published by S. Karger AG, Basel
www.karger.com/cnd
Spica et al.: Daratumumab for Treatment of Antibody-Mediated Rejection after ABO-
Incompatible Kidney Transplantation
141
immunoadsorption for the removal of circulating blood group antibodies. Although this ap-
proach prevented further decrease in allograft function and possibly graft loss, it did not in-
duce resolution of the rejection process due to the ongoing production of anti-A blood group
antibodies.
Based on the efficacy of daratumumab in the treatment of post-HSCT autoimmune hemo-
lytic anemia and pure red cell aplasia following ABO-incompatible HSCT, we hypothesized
that direct targeting and depletion of the antibody-producing plasma cells by daratumumab
could be a treatment option for our patient in order to avoid graft failure. Bortezomib has been
described for the treatment of refractory AMR in kidney transplantation in several cases [23,
24], which is a less directed approach to target plasma cells. However, we were reluctant to
give bortezomib because of the risk of severe peripheral neuropathy as a limiting side effect
in a patient who had been operated for spinal stenosis prior to kidney transplantation.
In fact, after the first dose of daratumumab, the suspected pathogenic IgM isohemagglu-
tinins continuously fell without any additional immunoadsorption. The rapid response could
be explained by the relatively short half-life of IgM antibodies of approximately 5 days. Al-
though we cannot prove the causality between daratumumab and the reduction in antibody
titers which could also be part of the natural course after ABO-incompatible kidney trans-
plantation the timely coincidence suggests the decrease in antibody titers to be a direct con-
sequence of daratumumab. Further, rising blood group antibody titers after transplantation
do not necessarily need to reflect subsequent allograft dysfunction or AMR, because allograft
tolerance of the blood group antibodies can be observed after ABO-incompatible kidney trans-
plantation. Even though a rise in blood group antibody titers can occur early in the post-trans-
plant course (i.e., from the first week ongoing), titers normally rise within 46 weeks after
transplantation, and “pure rebound” is typically not associated with graft dysfunction and his-
tologic signs of early active AMR [2527].
Moreover, there could be an influence of the complex antirejection therapy (steroid
pulses, anti-thymocyte globulins, eculizumab) administrated prior to daratumumab on the re-
duction of anti-A blood group antibodies and resolution of tissue damage. In a complex clinical
situation where a new therapeutic option is evaluated as a rescue therapy, it is impossible in
a single case to prove causality of the rescue therapy (i.e., therapeutic effect of a specific treat-
ment). One explanation in our case may be the fact that, despite those initial treatments, anti-
A IgM titers rose again two times immediately after discontinuation of immunoadsorption
and, in contrary, fell right after the first daratumumab administration without further immu-
noadsorption assuming a much greater effect of daratumumab on anti-A blood group anti-
body suppression. In addition, the level of blood group antibodies measured in the blood does
not necessarily reflect the burden of antibodies bound to the graft. However, the second allo-
graft biopsy performed on day 73 post-transplant showed no signs of acute rejection, includ-
ing no more deposition of IgM and complement C3c. Concerns about immunomodulatory ef-
fects of daratumumab occurred before infusion, as the drug targets CD38-expressing cells
through a variety of immune-mediated mechanisms [15]. Krejcik et al. [28] studied peripheral
blood and bone marrow of patients with relapsed/refractory myeloma which were treated
with daratumumab from two different monotherapy studies. They showed that daratumumab
depleted CD38-positive immune regulatory cells and promoted T-cell expansion, and demon-
strated further T-cell changes, which they highlighted as “skew of T-cell repertoire in multiple
myeloma” [28]. In transplantation, these immunomodulatory effects of the anti-CD38 drug
could represent additional mechanisms of interaction which may trigger cellular rejection
and, thus, act against the allograft. Thus, it is important to mention that the patient developed
no acute cellular rejection during the course and after treatment with daratumumab proven
Case Rep Nephrol Dial
DOI: 10.1159/000503951
© 2019 The Author(s). Published by S. Karger AG, Basel
www.karger.com/cnd
Spica et al.: Daratumumab for Treatment of Antibody-Mediated Rejection after ABO-
Incompatible Kidney Transplantation
142
by biopsy. Further, the medication was well tolerated by the patient, and no severe infections
were observed.
Conclusion
Our case report suggests that daratumumab may be a promising option as a rescue ther-
apy for therapy-refractory AMR in the context of ABO-incompatible kidney transplantation.
As large clinical trials evaluating new treatment regimens for AMR are unlikely to be per-
formed in the near future, case reports may be a more practical way to evaluate treatment
response. Furthermore, daratumumab may also be considered part of the preparative regi-
men before ABO-incompatible kidney transplantation with or without rituximab in order to
directly target blood group antibody-producing plasma cells with the potential to avoid im-
munoadsorption. Nevertheless, the immunomodulatory effects of daratumumab need to be
taken into account. To address this issue, further studies are warranted.
Statement of Ethics
The research was conducted in accordance with the World Medical Association Declara-
tion of Helsinki (https://www.wma.net/policies-post/wma-declaration-of-helsinki-ethical-
principles-for-medical-research-involving-human-subjects/). The participant gave written
informed consent for publication of this case report.
Disclosure Statement
The authors of this article declare no conflicts of interest as described by Karger Case Re-
ports in Nephrology and Dialysis.
Funding Sources
Daratumumab therapy was funded by Janssen-Cilag AG, Zug, Switzerland. P.H.-M. is sup-
ported by the Alfred und Erika Bär-Spycher-Stiftung.
Author Contributions
All authors contributed equally to the literature review and the text of the manuscript.
D.S., P.H.-M. and A.H. were responsible for the patient care and treatment.
References
1 Knoll G. Trends in kidney transplantation over the past decade. Drugs. 2008;68 Suppl 1:310.
2 Stegall MD, Park WD, Dean PG, Cosio FG. Improving long-term renal allograft survival via a road less traveled
by. Am J Transplant. 2011 Jul;11(7):13827.
3 Lamb KE, Lodhi S, Meier-Kriesche HU. Long-term renal allograft survival in the United States: a critical
reappraisal. Am J Transplant. 2011 Mar;11(3):45062.
Case Rep Nephrol Dial
DOI: 10.1159/000503951
© 2019 The Author(s). Published by S. Karger AG, Basel
www.karger.com/cnd
Spica et al.: Daratumumab for Treatment of Antibody-Mediated Rejection after ABO-
Incompatible Kidney Transplantation
143
4 Matas AJ, Smith JM, Skeans MA, Thompson B, Gustafson SK, Stewart DE, et al. OPTN/SRTR 2013 Annual Data
Report: kidney. Am J Transplant. 2015 Jan;15(S2 Suppl 2):134.
5 Stegall MD, Morris RE, Alloway RR, Mannon RB. Developing New Immunosuppression for the Next
Generation of Transplant Recipients: The Path Forward. Am J Transplant. 2016 Apr;16(4):1094101.
6 OʼConnell PJ, Kuypers DR, Mannon RB, Abecassis M, Chadban SJ, Gill JS, et al. Clinical Trials for
Immunosuppression in Transplantation: The Case for Reform and Change in Direction. Transplantation.
2017 Jul;101(7):152734.
7 Walsh L, Dinavahi R. Current unmet needs in renal transplantation: a review of challenges and therapeutics.
Front Biosci (Elite Ed). 2016 Jan;8(1):114.
8 Einecke G, Sis B, Reeve J, Mengel M, Campbell PM, Hidalgo LG, et al. Antibody-mediated microcirculation
injury is the major cause of late kidney transplant failure. Am J Transplant. 2009 Nov;9(11):252031.
9 Gaston RS, Cecka JM, Kasiske BL, Fieberg AM, Leduc R, Cosio FC, et al. Evidence for antibody-mediated injury
as a major determinant of late kidney allograft failure. Transplantation. 2010 Jul;90(1):6874.
10 Sellarés J, de Freitas DG, Mengel M, Reeve J, Einecke G, Sis B, et al. Understanding the causes of kidney
transplant failure: the dominant role of antibody-mediated rejection and nonadherence. Am J Transplant.
2012 Feb;12(2):38899.
11 Colvin RB, Smith RN. Antibody-mediated organ-allograft rejection. Nat Rev Immunol. 2005 Oct;5(10):807
17.
12 de Weerd AE, Betjes MG. ABO-Incompatible Kidney Transplant Outcomes: A Meta-Analysis. Clin J Am Soc
Nephrol. 2018 Aug;13(8):123443.
13 Archdeacon P, Chan M, Neuland C, Velidedeoglu E, Meyer J, Tracy L, et al. Summary of FDA antibody-
mediated rejection workshop. Am J Transplant. 2011 May;11(5):896906.
14 Davis S, Cooper JE. Acute antibody-mediated rejection in kidney transplant recipients. Transplant Rev
(Orlando). 2017 Jan;31(1):4754.
15 van de Donk NW, Usmani SZ. CD38 Antibodies in Multiple Myeloma: Mechanisms of Action and Modes of
Resistance. Front Immunol. 2018 Sep;9:2134.
16 Lonial S, Weiss BM, Usmani SZ, Singhal S, Chari A, Bahlis NJ, et al. Daratumumab monotherapy in patients
with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet.
2016 Apr;387(10027):155160.
17 Lokhorst HM, Plesner T, Laubach JP, Nahi H, Gimsing P, Hansson M, et al. Targeting CD38 with
Daratumumab Monotherapy in Multiple Myeloma. N Engl J Med. 2015 Sep;373(13):120719.
18 Costello C. An update on the role of daratumumab in the treatment of multiple myeloma. Ther Adv Hematol.
2017 Jan;8(1):2837.
19 Schuetz C, Hoenig M, Moshous D, Weinstock C, Castelle M, Bendavid M, et al. Daratumumab in life-
threatening autoimmune hemolytic anemia following hematopoietic stem cell transplantation. Blood Adv.
2018 Oct;2(19):25503.
20 Chapuy CI, Kaufman RM, Alyea EP, Connors JM. Daratumumab for Delayed Red-Cell Engraftment after
Allogeneic Transplantation. N Engl J Med. 2018 Nov;379(19):184650.
21 Oettl T, Halter J, Bachmann A, Guerke L, Infanti L, Oertli D, et al. ABO blood group-incompatible living donor
kidney transplantation: a prospective, single-centre analysis including serial protocol biopsies. Nephrol Dial
Transplant. 2009 Jan;24(1):298303.
22 Chapuy CI, Nicholson RT, Aguad MD, Chapuy B, Laubach JP, Richardson PG, et al. Resolving the
daratumumab interference with blood compatibility testing. Transfusion. 2015 Jun;55(6 Pt 2):154554.
23 Sureshkumar KK, Hussain SM, Marcus RJ, Ko TY, Khan AS, Tom K, et al. Proteasome inhibition with
bortezomib: an effective therapy for severe antibody mediated rejection after renal transplantation. Clin
Nephrol. 2012 Mar;77(3):24653.
24 Lee J, Kim BS, Park Y, Lee JG, Lim BJ, Jeong HJ, et al. The Effect of Bortezomib on Antibody-Mediated
Rejection after Kidney Transplantation. Yonsei Med J. 2015 Nov;56(6):163842.
25 Fehr T, Stussi G. ABO-incompatible kidney transplantation. Curr Opin Organ Transplant. 2012
Aug;17(4):37685.
26 Shah BV, Rajput P, Virani ZA, Warghade S. Baseline Anti-blood Group Antibody Titers and their Response to
Desensitization and Kidney Transplantation. Indian J Nephrol. 2017 May-Jun;27(3):1958.
27 Ishida H, Kondo T, Shimizu T, Nozaki T, Tanabe K. Postoperative rebound of antiblood type antibodies and
antibody-mediated rejection after ABO-incompatible living-related kidney transplantation. Transpl Int. 2015
Mar;28(3):28696.
28 Krejcik J, Casneuf T, Nijhof IS, Verbist B, Bald J, Plesner T, et al. Daratumumab depletes CD38+ immune
regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood. 2016
Jul;128(3):38494.
A. Holbro and P. Hirt-Minkowski contributed equally to this article
Case Rep Nephrol Dial
DOI: 10.1159/000503951
© 2019 The Author(s). Published by S. Karger AG, Basel
www.karger.com/cnd
Spica et al.: Daratumumab for Treatment of Antibody-Mediated Rejection after ABO-
Incompatible Kidney Transplantation
144
Fig. 1. Course of the 59-year-old patient treated with daratumumab for antibody-mediated acute rejection
after ABO-incompatible kidney transplantation. Daratumumab was administered at a dose of 16 mg per
kg of body weight in weekly intervals. MP, methylprednisolone (pulse doses between 250500 mg). Ecu-
lizumab (three infusions of 900 mg per week on days 12, 19, and 26, respectively). ATG, anti-human T-
lymphocyte globulins (i.e., totally seven infusions with a dosage between 24 mg/kg of body weight). Tac,
tacrolimus (goal trough level of 8−10 µg/L). Tac trough levels are indicated bi-weekly ±4 days and illus-
trate selected time points of measurement post-transplant. MMF, mycophenolate mofetil (twice daily 500
mg). Prednisone (3050 mg daily).
Case Rep Nephrol Dial
DOI: 10.1159/000503951
© 2019 The Author(s). Published by S. Karger AG, Basel
www.karger.com/cnd
Spica et al.: Daratumumab for Treatment of Antibody-Mediated Rejection after ABO-
Incompatible Kidney Transplantation
145
Fig. 2. Kidney transplant biopsy on day 7. a Conventional histology showed diffuse mild acute tubular
damage with widened tubular lumina and mild interstitial edema. PAS stain, original magnification ×100.
bd Immunofluorescence of cryosections demonstrated diffuse linear complement C4d staining along the
peritubular capillaries (b), but also a more granular deposition of IgM (c) and complement C3c (d), indi-
cating a very recent antibody interaction with the endothelium. Incubations for IgA and IgG were negative
(not shown). Indirect (b) and direct (c, d) immunofluorescence with FITC-labeled antibodies. Original
magnification ×200.
... Table 4 summarizes the mechanisms of action of the available therapies. Table 4. Mechanism of action of therapies used for dnDSA management [78,[86][87][88][89][90][91][92][93]. ...
... Alemtuzumab, an anti-CD52 monoclonal antibody that suppresses mature lymphocytes, has also been used in case reports of refractory rejection [92]. Finally, the use of other agents such as belimumab (anti-B-lymphocyte stimulator monoclonal antibody) or daratumumab (anti-CD38 monoclonal antibody) has been described in case reports with positive results [93,106]. However, the potential toxicity of these agents must be borne in mind before more solid evidence about their benefits is available. ...
Article
Full-text available
Antibodies directed against donor-specific human leukocyte antigens (HLAs) can be detected de novo after heart transplantation and play a key role in long-term survival. De novo donor-specific antibodies (dnDSAs) have been associated with cardiac allograft vasculopathy, antibody-mediated rejection, and mortality. Advances in detection methods and international guideline recommendations have encouraged the adoption of screening protocols among heart transplant units. However, there is still a lack of consensus about the correct course of action after dnDSA detection. Treatment is usually started when antibody-mediated rejection is present; however, some dnDSAs appear years before graft failure is detected, and at this point, damage may be irreversible. In particular, class II, anti-HLA-DQ, complement binding, and persistent dnDSAs have been associated with worse outcomes. Growing evidence points towards a more aggressive management of dnDSA. For that purpose, better diagnostic tools are needed in order to identify subclinical graft injury. Cardiac magnetic resonance, strain techniques, or coronary physiology parameters could provide valuable information to identify patients at risk. Treatment of dnDSA usually involves plasmapheresis, intravenous immunoglobulin, immunoadsorption, and ritxumab, but the benefit of these therapies is still controversial. Future efforts should focus on establishing effective treatment protocols in order to improve long-term survival of heart transplant recipients.
... 3 Schuetz et al. demonstrated successful daratumumab-treatment of refractory post-transplant autoimmune hemolytic anemia after allo-SCT in three patients. 12 Our uniquely ABO-as well as HLA antibodyincompatible case adds to the previously described reports of daratumumab use for ABMR therapy in kidney transplantation by Spica [13][14][15] We administered a regimen of only four doses of daratumumab subcutaneously instead of intravenously and decided to stop treatment based on the observed histopathological and clinical remission of ABMR. Application-associated toxicities or infections were not observed. ...
... 17 We cannot prove a definite causality between daratumumab and the observed reduction of anti-A titers and DSA levels albeit it is highly suggestive by the time course and limited response to PPH that initially resulted in a rebound of anti-A titers. Short half-life of IgM antibodies, an allograft tolerance for blood group antibodies, and a doubtful association of ABO-antibody titers with graft dysfunction and early active ABMR may also have contributed to the favorable clinical course, as also discussed by Spica et al. 13 However, we observed a lasting reduction of DSA and non-DSA HLA antibodies after daratumumab application, which cannot be explained by anti-A-immunoglobulinselective IA (Figure 2(b) and Table 2). However, other therapeutic regimens, such as more intense PPH treatment, could have been similarly effective cannot be excluded. ...
Article
Full-text available
We report a case of antibody-mediated rejection treated with the human CD38 monoclonal antibody daratumumab in a 58-year-old female patient with end-stage kidney disease due to autosomal dominant polycystic kidney disease who received an ABO- and human leukocyte antigen antibody-incompatible living donor kidney transplant. The patient experienced an episode of severe antibody-mediated rejection within the first week of transplantation. Blood-group-antibody selective immunoadsorption in combination with administration of four doses of daratumumab (each 1800 mg s.c.) led to a persistent decrease of ABO- and more interestingly donor-specific human leukocyte antigen antibody reactivity and resulted in clinical and histopathological remission with full recovery of graft function, which has remained stable until post-transplant day 212. This case illustrates the potential of targeting CD38 in antibody-mediated rejection.
... As such, novel strategies including therapies targeting plasma cells are being increasingly reported [6]. Daratumumab, an anti-CD38 antibody, successfully reduced the refractory DSAs in patients with B-acute lymphoblastic leukemia and AB0 incompatible solid organ transplantation [6,7]. The presence of CD38 on immature hematopoietic cells could theoretically lead to engraftment failure. ...
Article
Full-text available
Donor‐specific anti‐human leukocyte antigen (HLA) antibodies represent a main cause of primary graft failure specifically in the setting of haploidentical stem cell transplantation. Newer therapy strategies including daratumumab could overcome some of these limitations. We describe the case of a patient with refractory acute myeloid leukemia. A haploidentical allogeneic stem cell transplantation was therefore initiated. HLA‐antibodies testing revealed a high titer of donor‐specific antibodies. First desensitization therapy failed, resulting in primary graft failure. A second desensitization regimen including plasmapheresis, intravenous gammaglobulins, and daratumumab resulted in good engraftment. Daratumumab is a promising and effective desensitization option in high‐risk allo‐sensitized patients undergoing haploidentical stem cell transplantation.
... Daratumumab is an IgG 1-kappa monoclonal antibody that binds to CD38 transmembrane glycoprotein and induces plasma cell death. It was first studied for the treatment of multiple myeloma [64]. It is dosed as an IV infusion of 16 mg/kg weekly for 8 weeks. ...
Article
Full-text available
Purpose of Review The goal of this paper is to review the latest advances in antibody-mediated rejection with regards to lung transplantation. We review definitions, mechanisms, pathology, and the most recent therapeutics that have been trialed for treating AMR. We hope to answer questions regarding diagnostic ambiguity that previously surrounded AMR and hope to shed light to the latest treatments which have shown promise in recent clinical trials. Recent Findings We review the benefits of single bead Luminex assays, and how the implementation of these assays has improved detection for donor specific antibodies, a key component in the diagnosis of AMR. We also explore other serum markers that may be beneficial in the diagnosis of AMR such as p-S5RP, and cell- free DNA. Summary There are new treatment options to consider such as the IL6 inhibitors Tocilizumab and clazakizumab, as well as tyrosine kinase inhibitors such as felzartamab or inflimidase, all of which have shown some promise for treating AMR.
... Daratumumab is a monoclonal antibody against CD38 which has shown promising results in decreasing anti-HLA antibodies in AMR [86,87] and has been proposed as an agent for desensitization and treatment of AMR. However, more concrete evidence is needed before its acceptance in clinical practice. ...
Article
Full-text available
With increasing knowledge of immunologic factors and with the advent of potent immunosuppressive agents, the last several decades have seen significantly improved kidney allograft survival. However, despite overall improved short to medium-term allograft survival, long-term allograft outcomes remain unsatisfactory. A large body of literature implicates acute and chronic rejection as independent risk factors for graft loss. In this article, we review measures taken at various stages in the kidney transplant process to minimize the risk of rejection. In the pre-transplant phase, it is imperative to minimize the risk of sensitization, aim for better HLA matching including eplet matching and use desensitization in carefully selected high-risk patients. The peri-transplant phase involves strategies to minimize cold ischemia times, individualize induction immunosuppression and make all efforts for better HLA matching. In the post-transplant phase, the focus should move towards individualizing maintenance immunosuppression and using innovative strategies to increase compliance. Acute rejection episodes are risk factors for significant graft injury and development of chronic rejection thus one should strive for early detection and aggressive treatment. Monitoring for DSA development, especially in high-risk populations, should be made part of transplant follow-up protocols. A host of new biomarkers are now commercially available, and these should be used for early detection of rejection, immunosuppression modulation, prevention of unnecessary biopsies and monitoring response to rejection treatment. There is a strong push needed for the development of new drugs, especially for the management of chronic or resistant rejections, to prolong graft survival. Prevention of rejection is key for the longevity of kidney allografts. This requires a multipronged approach and significant effort on the part of the recipients and transplant centers.
Article
A BSTRACT Daratumumab is a monoclonal antibody; anti-CD38 is used for the treatment of multiple myeloma and other hematological disorders. Daratumumab, binds to CD 38 on red blood cells (RBCs), interfering with pretransfusion testing. This interference can lead to cross match incompatibility and panreactive indirect Antiglobulin tests (IAT).Panreactive IAT can mask presence of underlying clinically significant alloantibodies, especially in patient with history of multiple blood transfusions. This can lead to delays in transfusion, increased risk of transfusion reaction and potential risk for transfusion recipients. Daratumumab induced potential serological interference needs to be resolved by using appropriate methodologies like dithiotheritol (DDT) and enzymatic treatment of reagents or donors cells to overcome these incompatibilities. Effective communication between clinical practitioners and blood centre before starting daratumumab therapy can prevent unnecessary workload and delay in transfusion. Here, we describe the case of interference in prretransfuison testing caused by Daratumumab in the red cell compatibility testing of a patient with severe aplastic anemia and strongly positive DSA levels.
Article
Full-text available
CD38 antigen is a glycoprotein that found on the surface of several immune cells, and this property makes its monoclonal antibodies have the effect of targeted elimination of immune cells. Therefore, the CD38 monoclonal antibody (such as daratumumab, Isatuximab) becomes a new treatment option for membranous nephropathy, lupus nephritis, renal transplantation, and other refractory kidney diseases. This review summarizes the application of CD38 monoclonal antibodies in different kidney diseases and highlights future prospects.
Article
Full-text available
b> Introduction: The transplantation of highly sensitized patients remains a major obstacle. Immunized patients wait longer for a transplant if not prioritized, and if transplanted, their transplant outcome is worse. Case Presentation: We report a successful AB0- and HLA-incompatible living donor kidney transplantation in a 35-year-old female patient with systemic lupus erythematosus (SLE) and antiphospholipid syndrome. The patient had a positive T- and B-cell complement-dependent cytotoxicity (CDC) crossmatch and previous graft loss due to renal vein thrombosis. We treated the patient with intravenous immunoglobulins, rituximab, horse anti-thymocyte globulin, daratumumab, and imlifidase, besides standard immunosuppression. All IgG antibodies were sensitive to imlifidase treatment. Besides donor-specific HLA antibodies, anti-dsDNA antibodies and antiphospholipid antibodies were cleaved. The patient initially had delayed graft function. Two kidney biopsies (day 7 and day 14) revealed acute tubular necrosis without signs of HLA antibody-mediated rejection. On posttransplant day 30, hemodialysis was stopped, and creatinine levels declined over the next weeks to a baseline creatinine of about 1.7 mg/dL after 12 months. Conclusion: In this case, a novel multimodal treatment strategy including daratumumab and imlifidase enabled successful kidney transplantation for a highly immunized patient with antiphospholipid antibodies.
Article
Full-text available
Daratumumab, a human IgG1κ monoclonal antibody targeting CD38, is used to treat multiple myeloma. We describe successful treatment with daratumumab in a case of treatment-refractory pure red-cell aplasia after ABO-mismatched allogeneic stem-cell transplantation. The patient was a 72-year-old man with the myelodysplastic syndrome who received a transplant from an HLA-matched, unrelated donor with a major ABO incompatibility (blood group A in the donor and blood group O in the recipient). The patient had persistent circulating anti-A antibodies and no red-cell recovery 200 days after transplantation. Standard treatments had no effect. Within 1 week after the initiation of treatment with daratumumab, he no longer required transfusions.
Article
Full-text available
MM cells express high levels of CD38, while CD38 is expressed at relatively low levels on normal lymphoid and myeloid cells, and in some non-hematopoietic tissues. This expression profile, together with the role of CD38 in adhesion and as ectoenzyme, resulted in the development of CD38 antibodies for the treatment of multiple myeloma (MM). At this moment several CD38 antibodies are at different phases of clinical testing, with daratumumab already approved for various indications both as monotherapy and in combination with standards of care in MM. CD38 antibodies have Fc-dependent immune effector mechanisms, such as complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP). Inhibition of ectoenzymatic function and direct apoptosis induction may also contribute to the efficacy of the antibodies to kill MM cells. The CD38 antibodies also improve host-anti-tumor immunity by the elimination of regulatory T cells, regulatory B cells, and myeloid-derived suppressor cells. Mechanisms of primary and/or acquired resistance include tumor-related factors, such as reduced cell surface expression levels of the target antigen and high levels of complement inhibitors (CD55 and CD59). Differences in frequency or activity of effector cells may also contribute to differences in outcome. Furthermore, the microenvironment protects MM cells to CD38 antibody-induced ADCC by upregulation of anti-apoptotic molecules, such as survivin. Improved understanding of modes of action and mechanisms of resistance has resulted in rationally designed CD38-based combination therapies, which will contribute to further improvement in outcome of MM patients.
Article
Full-text available
In recent years, immunological barriers historically considered as absolute contraindications to transplantation are being reevaluated. One such barrier is the ABO blood group incompatibility. With better understanding of immunological mechanisms and effective various regimens for controlling it, ABO-incompatible (ABO-I) kidney transplantation is now being performed with increasing frequency. For good outcome, most important is to achieve and maintain low anti-blood group antibody titers (ABGATs). Twenty-two patients with ABO-I donors have been studied. The anti-A and anti-B antibody titers (IgG and IgM) were estimated by column agglutination technology using Automated Ortho BioVue System. For desensitization, pretransplant plasmapheresis and/or immunoadsorption and rituximab were used. ABGAT was determined before transplant and periodically after transplant. It was observed that one-third of the patients have low baseline ABGAT. In these cases with low ABGAT, transplant can be performed without any desensitization. In those with titers <1:256, rituximab (two doses of 200 mg weekly) and 3-6 sessions of plasmapheresis can bring down titers to <1:32. In those with titers >1:256, immunoadsorption may be used from the beginning to reduce ABGAT. After transplant, the titers drop to <1:8 in majority. Rise in titers to >1:64 require close observation and biopsy. If there is evidence of antibody-mediated rejection, treatment should be promptly started. Rise in titers 4-6 weeks after transplant is not associated with any graft dysfunction, and hence not of any clinical significance. © 2017 Indian Journal of Nephrology | Published by Wolters Kluwer - Medknow.
Article
Full-text available
Monoclonal antibodies (mAbs) have emerged as a promising new drug class for the treatment of multiple myeloma (MM). Daratumumab (DARA), a CD38 mAb, has demonstrated safety, tolerability and activity in a range of clinical trials, both as monotherapy and in combination strategies for MM. The favorable efficacy results in heavily pretreated patients with advanced MM have provided the rationale for the investigation of DARA in a number of ongoing and future phase II and III trials. The general tolerability of mAbs has allowed for widespread investigation and use of DARA among a variety of MM patients, however their use requires special consideration. Infusion-related reactions (IRRs), interference with blood compatibility assays and response assessments are all unique factors related to the use of DARA. This review provides an update of the results from the DARA clinical trials conducted to date, its future plans for investigation, and practical management considerations for the use of DARA in daily practice.
Article
New-onset autoimmune hemolytic anemia (AIHA) occurs in 2% to 6% of pediatric patients post-hematopoietic stem cell transplantation (HSCT) and is a significant complication. Incomplete immune recovery following HSCT may predispose to immune dysregulation including autoimmune cytopenias. We describe an innovative therapy for AIHA refractory to proteasome inhibition. In potentially life-threatening AIHA in the context of HSCT, daratumumab may be an effective rescue therapy.
Article
Background and objectives: ABO blood group-incompatible kidney transplantation is considered a safe procedure, with noninferior outcomes in large cohort studies. Its contribution to living kidney transplantation programs is substantial and growing. Outcomes compared with center-matched ABO blood group-compatible control patients have not been ascertained. Design, setting, participants, & measurements: Comprehensive searches were conducted in Embase, Medline, Cochrane, Web-of-Science, and Google Scholar. Meta-analyses Of Observational Studies in Epidemiology study guidelines for observational studies and Newcastle Ottawa bias scale were implemented to assess studies. Meta-analysis was performed using Review Manager 5.3. A subgroup analysis on antibody removal technique was performed. Results: After identifying 2728 studies addressing ABO-incompatible kidney transplantation, 26 studies were included, describing 1346 unique patients who were ABO-incompatible and 4943 ABO-compatible controls. Risk of bias was low (all studies ≥7 of 9 stars). Baseline patient characteristics revealed no significant differences in immunologic risk parameters. Statistical heterogeneity of studies was low (I2 0% for graft and patient survival). One-year uncensored graft survival of patients who were ABO-incompatible was 96% versus 98% in ABO-compatible controls (relative risk, 0.97; 95% confidence interval, 0.96 to 0.98; P<0.001). Forty-nine percent of reported causes of death in patients who were ABO-incompatible were of infectious origin, versus only 13% in patients who were ABO-compatible (P=0.02). Antibody-mediated rejection (3.86; 95% confidence interval, 2.05 to 7.29; P<0.001), severe nonviral infection (1.44; 95% confidence interval, 1.13 to 1.82; P=0.003), and bleeding (1.92; 95% confidence interval, 1.36 to 2.72; P<0.001) were also more common after ABO-incompatible transplantation. Conclusions: ABO-incompatible kidney transplant recipients have good outcomes, albeit inferior to center-matched ABO-compatible control patients.
Article
Currently trials of immunosuppression in transplantation are in decline because their objectives remain focused on improving acute rejection rates and graft survival in the first 12 months. With 1 year renal graft survival rates of greater than 90% the best that can be hoped for is noninferiority trial outcomes compared to current standard of care. Current trial design is not leading to novel therapies improving long term outcomes and safety, and hence important unmet clinical needs in transplantation remain unanswered. Issues that need to be addressed include but are not limited to: prevention of subclinical rejection in the first year, better 5 and 10 year graft outcomes, more effective treatment for high immunological risk and sensitized (including DSA) patients, immunosuppressive combinations that are better tolerated by patients with fewer side effects and less morbidity and mortality. In September 2015 the Transplantation Society convened a group of transplant clinical trial experts to address these problems. The aims were to substantially realign the priorities of clinical trials for renal transplant immunosuppression with the current unmet needs and to propose new designs for clinical trials for transplant immunosuppression. Moving forward, the transplant community needs to provide trial data that will identify superior treatment options for patient subgroups and allow new agents to be evaluated for efficacy and safety and achieve timely regulatory approval. Trial designs for new transplant immunosuppression must be intelligently restructured in order to ensure that short- and long-term clinical outcomes continue to improve.
Article
Antibody-mediated rejection has now been recognized as one of the most important causes of graft loss. Transplantation across HLA barriers and non-adherence can result in acute antibody-mediated rejection, which is associated with particularly worse graft outcomes. New technologies, including genomic studies and assays to detect and define donor-specific antibodies, have provided important insights into the pathophysiology and diagnosis of acute antibody-mediated rejection but have engendered many questions about the clinical application of these tests in the prognosis and prevention of this protean disease process. In this article, we review the pathophysiology of acute antibody-mediated rejection, the evolving diagnostic criteria, and specific challenges related to its prognosis, treatment, and prevention.
Article
Key Points CD38-expressing immunosuppressive regulatory T and B cells and myeloid-derived suppressor cells were sensitive to daratumumab treatment. Cytotoxic T-cell number, activation, and clonality increased after daratumumab treatment in heavily pretreated relapsed and refractory MM.
Article
While has been considerable progress in the short-term outcomes following renal transplantation over the last several decades, minimal gains have been made with regards to long-term graft function and patient survival (1). The lack of long-term gains has been attributed to factors such as antibody mediated rejection (AMR), chronic allograft nephropathy (CAN), and toxicity to the allograft secondary to immunosuppression. Ischemia reperfusion injury (IRI) is also thought to contribute to poor long-term graft function, and its impact on patient and graft outcomes will likely expand with the increasing use of marginal kidneys secondary to organ shortages. While patient survival remains far below that of the general population, the causes of death have evolved in recent years with decreases in the rate of death from cardiovascular disease and infection, and increases secondary to malignancy (2), which are largely attributable to the potency of modern immunosuppression. As such, the development of novel therapies which can prevent delayed graft function (DGF), minimize AMR, while simultaneously reducing toxicity is vital to the improvement of long-term graft and patient outcomes.