A literature review of the use of daratumumab in non-neoplastic immunologic disorders
Review Article

A literature review of the use of daratumumab in non-neoplastic immunologic disorders

Jennifer M. Jones1 ORCID logo, Minh-Ha Tran2

1Department of Pathology, University of Michigan, Ann Arbor, MI, USA; 2Department of Pathology and Laboratory Medicine, University of California, Irvine, Orange, CA, USA

Contributions: (I) Conception and design: Both authors; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: Both authors; (V) Data analysis and interpretation: Both authors; (VI) Manuscript writing: Both authors; (VII) Final approval of manuscript: Both authors.

Correspondence to: Jennifer M. Jones, MD. Department of Pathology, University of Michigan, 2F355B UH, 1500 E. Medical Center Dr., Ann Arbor, MI 48109-5054, USA. Email: jonjm@med.umich.edu.

Background and Objective: Daratumumab, a novel anti-CD38 monoclonal antibody therapy approved for multiple myeloma, has seen increased use in the treatment of non-neoplastic conditions. This literature review summarizes the published experience with daratumumab in non-neoplastic immunologic disorders.

Methods: A literature search was conducted in PubMed utilizing the following MESH terms: (Daratumumab) AND (immune) with added restrictions of humans, English language, and date range January 1, 2015 to November 1, 2023. Articles were first screened by title and abstract. Preclinical animal studies, in vitro experiments, reviews, or reports with non-immunologic use intention were excluded. Instances of uncertainty were resolved through full manuscript review and discussion between authors. Extracted datapoints included indication, prior lines of treatment, days to initiation of daratumumab, its dose and route, and response to daratumumab. Patient demographics and infusion reactions to daratumumab were recorded if available. Extracted data were analyzed according to diagnostic group (wherever 3 or more individuals had the same daratumumab use indication) and utilizing descriptive statistics. Nonparametric data are presented as medians with interquartile ranges.

Key Content and Findings: The literature search yielded 192 articles for consideration. Following review, 50 reports, encompassing 99 patients met inclusion criteria. Daratumumab was most used in the following conditions: post-transplant autoimmune hemolytic anemia (22, 22%), post-transplant pure red cell aplasia (11, 11%), post-transplant autoimmune cytopenias (9, 9%), thrombotic thrombocytopenic purpura (9, 9%), systemic lupus erythematosus (8, 8%), immune thrombocytopenia (7, 7%), autoimmune encephalitis (6, 6%), and organ transplant rejection (5, 5%). Prior to daratumumab, patients received a median of 4 lines [interquartile range (IQR), 2 to 5 lines] of therapy. Complete and partial responses were reported in 58% and 31% of cases, respectively. Median time to response with daratumumab was 28 days (IQR, 14 to 61 days), and responses were durable (median 270 days; IQR, 123 to 365 days). Mild infusion reactions occurred in 11 cases (11%). No severe allergic reactions or deaths due to daratumumab were reported.

Conclusions: Our comprehensive review highlights the efficacy, durability of response, and safety of daratumumab in several refractory non-neoplastic conditions. Future investigation may define a role for daratumumab earlier in the treatment hierarchy for select antibody-mediated disorders.

Keywords: Daratumumab; antibody-mediated disorders; refractoriness


Received: 23 August 2024; Accepted: 12 December 2024; Published online: 29 December 2024.

doi: 10.21037/aob-24-23


Introduction

Non-neoplastic immune disorders span multiple disciplines—from hematology to neurology to rheumatology and beyond. Despite heterogeneity in presentation, these conditions share the feature of being caused by humoral immune activity. Autoimmune conditions result from failure of self-tolerance, where self-antigens become the target of immune responses (1). Diagnosis of these conditions often requires identification of causative antibodies. For example, the presence of anti-double stranded DNA aides in the diagnosis of systemic lupus erythematosus (SLE), and antibodies against the acetylcholine receptor signal a diagnosis of myasthenia gravis (2,3). Unlike autoimmune disorders, some antibody-mediated conditions, such as red blood cell (RBC) alloimmunization and antibody-mediated organ transplant rejection, arise from appropriate immune identification of and response to non-self-antigens but are no less unwanted (4,5).

Options to manage non-neoplastic immune disorders target all known aspects of the humoral immune response. There are no known methods to prevent initial self-antigen recognition in autoimmune disorders, and the only definitive method to prevent alloimmunization in transplant and transfusion recipients is to avoid exposure to non-identical or compatible antigens, which is not always feasible. Following onset of an antibody-mediated condition, traditional management approaches have focused on minimizing the impact of these antibodies via mechanical removal with plasmapheresis, antibody cleavage, and antibody effector activity blockade (e.g., with Fc receptor inhibition) (6-10). Efforts are also made to reduce antibody production through lymphocyte depletion. Although antibody-secreting plasma cells arise from B-cells, B- and T-cells are often the target for these therapies because helper T-cell co-stimulation can assist B-cell affinity maturation in lymphoid tissues (11,12). Therapies such as systemic corticosteroids, calcineurin inhibitors, mTOR inhibitors, antimetabolites, anti-thymocyte globulin, and traditional chemotherapies have been used to facilitate lymphocyte depletion (2,6,10,13,14). Disadvantages to all these approaches include their non-specific effects which can cause generalized immunosuppression and increased susceptibility to infections, non-infectious adverse drug effects, and, for some therapies, high ancillary resource requirements. For example, plasmapheresis requires specialized equipment and trained personnel which may not be universally available (15).

Monoclonal antibody (MoAb) therapies have seen increasing use in the treatment of non-neoplastic immune disorders because they (I) exhibit specific activity to immune receptors or proteins; (II) are easy to administer; and (III) have a favorable safety profile (16-18). Rituximab, an anti-CD20 MoAb therapy, revolutionized the treatment landscape of several antibody-mediated autoimmune and alloimmune disorders. In combination with other immunosuppressive agents, rituximab effectively removes CD20+ B-cells ideally before they have had the opportunity to mature into antibody-secreting plasma cells (19). However, mechanisms of resistance to rituximab have been described, and some patients exhibit persistent antibody-mediated pathology despite receiving rituximab (20). It has been theorized that antibodies produced by long-lived CD38+ plasma cells (LLPCs) play a prominent role in antibody-mediated disorders that are refractory to rituximab and other traditional immunosuppressive therapies (21-23). LLPCs do not express CD20 and are not susceptible to anti-CD20 therapy. Further bone marrow LLPCs exist in niches that promote long-term survival and durability (24).

Daratumumab, a novel anti-CD38 monoclonal IgG1 kappa therapy, has emerged as a promising therapy for refractory antibody-mediated disorders, where traditional immunosuppression and anti-CD20 therapies have been ineffective and persistent antibody production is thought due to CD38+ plasma cells (25). Daratumumab received accelerated Food and Drug Administration (FDA) approval in 2015 for the treatment of multiple myeloma (26). However, it has been increasingly applied “off-label” to treat refractory, non-neoplastic conditions. Clinical trials demonstrating the efficacy of daratumumab in these conditions have not been performed in large part because many are rare, rendering trials infeasible. Several case reports and case series have been published detailing the use of daratumumab in non-neoplastic conditions, though few current reviews have summarized these studies. Our objective was to highlight disease states with the most evidence supporting daratumumab use and to identify areas where daratumumab may be efficacious but require additional investigation. We present this article in accordance with the Narrative Review reporting checklist (available at https://aob.amegroups.com/article/view/10.21037/aob-24-23/rc).


Methods (see Table 1)

Table 1

Search details

Items Specification
Date of search November 22, 2023
Database searched PubMed
Search terms used (Daratumumab) AND (immune)
Restrictions: human, English language
Timeframe January 1, 2015 to November 1, 2023
Exclusion criteria Preclinical animal studies, in vitro experiments, reviews, or reports with non-immunologic use intention
Selection process J.M.J. and M.H.T. divided the list and screened titles and abstracts. Instances of uncertainty were resolved through discussion
Additional considerations Date range was selected to include all relevant literature since the FDA approval of daratumumab in 2015

FDA, Food and Drug Administration.

A literature search was conducted in PubMed utilizing the following MESH terms: (Daratumumab) AND (immune) with added restrictions of humans, English language, and date range January 1, 2015 to November 1, 2023. Articles were first screened by title and abstract. Preclinical animal studies, in vitro experiments, reviews, or reports with non-immunologic use intention were excluded. Instances of uncertainty were resolved through full manuscript review and discussion between authors. During full-text review of retained articles, additional reports were identified (i.e., bibliographic search). Extracted datapoints included indication, therapeutic agents used prior to daratumumab (referred to as “prior lines of therapy”), days to initiation of daratumumab, its dose and route, and response to daratumumab. Studies without these core datapoints were excluded. Responses to daratumumab were graded as complete response (CR), partial response (PR), or no response (NR) based on treatment response criteria for each condition as described by national societies, consensus guidelines, or foundational clinical trials (Table S1). Patient demographics (sex and age) and infusion reactions to daratumumab were recorded if available. Extracted data were analyzed according to diagnostic group (wherever 3 or more individuals had the same daratumumab use indication) and utilizing descriptive statistics. Nonparametric data are presented as medians with interquartile ranges.


Results

The initial search returned 192 reports of which 36 reports comprising 95 cases met initial inclusion criteria. Adjudication between authors led to exclusion of one report entailing 14 cases of pediatric nephrotic syndrome (27) leaving 35 reports comprising 81 cases. Of these cases, 2 reports with 3 duplicate cases (28,29) were identified and removed leaving 33 reports and 78 cases. Through bibliographic search, 17 additional reports comprising 21 cases were found for a total of 50 reports and 99 cases (Figure 1). Sex was reported for 83/99 (84%) and age for 95/99 (96%); 43/83 (52%) were female and the mean age was 36 years (range, 0.8–82 years).

Figure 1 Flowchart of article selection.

Published experience with daratumumab spanned several antibody-mediated conditions. Of included cases, 92/99 (93%) cases fell into 12 diagnostic groups containing 3 or more patients (Table 2): autoimmune hemolytic anemia (AIHA) (30-39), autoimmune cytopenias (AIC) (36,37,40,41), acquired hemophilia A (Acq Hem A) (11), myositis/myopathy/myasthenia (M/M/M) (42-44), immune thrombocytopenia (ITP) (37,45-47), cold agglutinin disease (CAD) (48-51), SLE (52,53), antibody mediated rejection (ABMR) (54-56), thrombotic thrombocytopenic purpura (TTP) (57-59), hemophilia A with inhibitors (60), pure red cell aplasia (PRCA) (61-70), and encephalitis (29,44,71). Seven of 99 cases fell into 5 diagnostic groups that contained 2 patients or less: RBC alloimmunization (72), human leukocyte antigen (HLA) alloimmunization (55,72), post-transplant hepatitis (73), antiphospholipid syndrome (74), and acquired von Willebrand disease (75) (Table 2).

Table 2

Basic demographics and daratumumab responses for all included patients

Disease # Cases Female Age (years), mean [range] Dara: CR/PR/NR
(Post Tx) AIHA 22 9/14 (64%) 20 [0.8–60] 10/8/4
Post Tx PRCA 11 5/11 (46%) 47 [14–77] 10/1/0
(Post Tx) AIC 9 1/2 (50%) 15 [0.8–60] 6/1/2
TTP 9 7/9 (78%) 49 [31–71] 7/1/1
SLE 8 8/8 (100%) 40 [20–61] 4/3/1
ITP 7 3/6 (50%) 38 [4–82] 4/0/3
Autoimmune encephalitis 6 2/6 (33%) 42[16–69] 2/4/0
Organ transplant rejection 5 1/5 (20%) 45 [29–59] 3/2/0
Acquired hemophilia A 4 1/4 (25%) 53 [32–73] 4/0/0
Cold agglutinin disease 4 1/4 (25%) 58 [48–69] 1/3/0
Myositis/myopathy/myasthenia 4 0/4 39 [19–57] 2/2/0
Hemophilia A with Inhibitors 3 0/3 18 [16–21] 0/3/0
RBC alloimmunization 2 2/2 (100%) 16 [14–17] 2/0/0
HLA alloimmunization 2 2/2 (100%) 44 [19–69] 1/1/0
Post Tx hepatitis 1 0/1 16 0/1/0
Antiphospholipid syndrome 1 1/1 (100%) 21 0/1/0
Acquired VWD 1 0/1 66 1/0/0
Total 99 43/83 (52%) 36 [0.8–82] 57/31/11

CR, complete response; PR, partial response; NR, no response; Post Tx, post transplant; AIHA, autoimmune hemolytic anemia; PRCA, pure red cell aplasia; AIC, autoimmune cytopenia; TTP, thrombotic thrombocytopenic purpura; SLE, systemic lupus erythematosus; ITP, immune thrombocytopenia; RBC, red blood cell; HLA, human leukocyte antigen; VWD, von Willebrand disease.

Patients within the 12 diagnostic groups containing 3 or more patients received a median of 4 lines [interquartile range (IQR), 2 to 5 lines] of therapy prior to daratumumab. Prior lines of therapy were loosely grouped according to mechanism of action (Table 3). There were 370 total therapeutic use instances recorded (excluding daratumumab); 253 (68.4%) antibody/B-cell/plasma-cell directed therapeutics (Ab/B/PC), 45 (12.2%) immunosuppressants/anti-rejection agents (IS/AR), 28 (7.6%) disease specific and supportive therapies (Dz Spec), 22 (5.9%) antineoplastic/antimetabolites (AN/AM), 9 (2.4%) other MoAbs, 8 (2.2%) other agents, 5 (1.4%) kinase inhibitors (KI) (Table 4). Within the 12 diagnostic groups, the broadest array of therapeutic categories attempted were in treatment of AIHA, AIC, Acq Hem A, and M/M/M at 6/7; followed by ITP and CAD at 5/7, SLE at 4/7, ABMR and TTP at 3/7, and hemophilia A with inhibitors, post transplant PRCA, and encephalitis at 2/7 each (Table 4).

Table 3

Categorization of therapeutic agents and utilization rate per category

Category Drugs Utilization rate*
AN/AM Methotrexate, cyclophosphamide, bendamustine, vincristine 22/370 (5.9%)
IS/AR Azathioprine, mycophenolate mofetil, sirolimus, cyclosporin, tacrolimus, basiliximab, antithymocyte globulin 45/370 (12.2%)
MoAb Alemtuzumab, abatacept, eculizumab 9/370 (2.4%)
KI Ibrutinib, fosfamatinib, zanabrutinib, trametinib 5/370 (1.4%)
Other agents Danazol, hydroxychloroquine, lenalidomide 8/370 (1.4%)
Dz Spec Immune tolerance induction, factor VIII bypassing agents, caplacizumab, thrombopoietin-receptor agonists, erythroid stimulating agents 28/370 (7.6%)
Ab/B/PC Corticosteroids, IVIG, therapeutic plasma exchange, splenectomy, rituximab, ofatumumab, belimumab, bortezomib 253/370 (68.4%)

*, total number of agents used under each category for all 99 patients. Total percentage 100.1% due to rounding. AN/AM, antineoplastic/antimetabolite; IS/AR, immunosuppressants/anti-rejection agents; MoAb, other monoclonal antibodies; KI, kinase inhibitors; Dz Spec, disease specific and supportive; Ab/B/PC, antibody/B-cell/plasma-cell directed therapeutics (daratumumab excluded); IVIG, intravenous immunoglobulin.

Table 4

Therapies utilized prior to daratumumab by diagnostic group

Disease category AN/AM IS/AR MoAb KI Other Dz Spec Ab/B/PC
AIHA (n=22) 4 17 5 2 0 5 59
Post Tx AIC (n=9) 1 3 1 0 1 3 22
Acquired hemophilia A (n=4) 4 2 0 1 1 1 7
Myositis/myopathy/myasthenia (n=4) 2 3 1 1 1 0 15
Immune thrombocytopenia (n=7) 1 3 0 0 3 5 19
Cold agglutinin disease (n=4) 3 0 0 1 1 1 10
Systemic lupus erythematosus (n=8) 6 10 0 0 1 0 21
Antibody mediated rejection (n=5) 0 4 2 0 0 0 15
Thrombotic thrombocytopenic purpura (n=9) 0 3 0 0 0 4 36
Post Tx PRCA (n=11) 0 0 0 0 0 3 15
Hemophilia A with inhibitors (n=3) 0 0 0 0 0 4 2
Autoimmune encephalitis (n=6) 1 0 0 0 0 0 24

For each patient in the category, use of an agent was recorded and totaled. AN/AM, antineoplastic/antimetabolite; IS/AR, immunosuppressants/anti-rejection agents; MoAb, other monoclonal antibodies; KI, kinase inhibitors; Other, other agents; Dz Spec, disease specific and supportive; Ab/B/PC, antibody/B-cell/plasma-cell directed therapeutics (daratumumab excluded); Post Tx, post transplant; AIHA, autoimmune hemolytic anemia; PRCA, pure red cell aplasia; AIC, autoimmune cytopenia.

Most reported cases showed a positive response to daratumumab. The overall CR, PR, and NR response rates were 57/99 (58%), 31/99 (31%), and 11/99 (11%), respectively (Table 2). Any response therefore occurred in 88/99 (89%). Time to response could be assessed in 77/88 (88%) of responders and occurred at a median timepoint in days of 32 (IQR, 14 to 60). Durability of response was assessable in 81/88 (92%) of responders. Median duration of response was 270 days (IQR, 153 to 365 days). Presence or absence of disease recurrence could be assessed in 86/99 (87%) patients. Absence of disease recurrence during follow up was noted in 73/86 (85%), with recurrences in 12/86 (14%). Presence or absence of daratumumab-related acute infusion reactions could be assessed in 96/99 (97%) patients, of whom 11/96 (12%) experienced infusion reactions, which were generally mild. The delayed complication of hypogammaglobulinemia was specifically attributed to daratumumab in 12/99 (12%) patients (Table 5).

Table 5

Summary of infusion/late reactions

Report Dx Age (years)/sex Dose (mg/kg)/route Response Infusion reaction symptoms Delayed complications
Driouk (36) Post Tx Evan’s 6/male 16/IV NR Bronchial obstruction and vomiting None
Post Tx AIHA 11/female 16/IV CR Nasal congestion and bronchial obstruction with first infusion only None
Khandelwal (37) XIAP Def s/p HSCT → AIHA 12/NR 16/IV CR Headache and emesis with first dose None
CTLA 4 Def s/p HSCT → AIHA 15/NR 16/IV CR Hypertension 24 hours later None
IPEX → AIN, AIT, AIHA 18/NR 16/IV PR Scratchy throat with first dose None
CGD s/p HSCT → AIHA 23/NR 16/IV NR Hypertension and chest pain None
Scheibe (44) Autoimmune Encephalitis 16/male 16/IV CR None Hypogammaglobulinemia (6 total doses)
27/female 16/IV CR None Hypogammaglobulinemia (4 total doses)
59/male 16/IV PR None Hypogammaglobulinemia (8 total doses)
60/male 16/IV PR None Hypogammaglobulinemia (13 total doses)
69/male 16/IV PR None Hypogammaglobulinemia (8 total doses)
CIDP/SLONM 52/male 16/IV CR None Hypogammaglobulinemia (20 total doses)
Seronegative MG 57/male 16/IV PR None Hypogammaglobulinemia (20 total doses)
Vernava (45) ITP 54/male 16/IV NR Dyspnea, agitation steroid flush None
Mohamed (49) CAD 69/female 16/IV CR Grade I fatigue Hypogammaglobulinemia
Zaninoni (51) CAD 59/male 16/IV PR None Hypogammaglobulinemia (19 total doses)
Roccatello (53) Lupus nephritis 20/female 16/IV CR Facial flushing None
Doberer (54) Renal transplant rejection 49/male 16/IV PR Allergic rhinitis (mild) with first dose Hypogammaglobulinemia (total doses NR)
Asawapanumas (64) Post-HSCT PRCA 49/male 16/IV CR Manageable infusion reactions … were observed None
Martino (68) Post-HSCT PRCA 62/female 16/IV CR None Hypogammaglobulinemia (2 total doses)
77/female 16/IV CR None Hypogammaglobulinemia (2 total doses)

Post Tx, post transplant; HSCT, hematopoietic stem cell transplant; AIHA, autoimmune hemolytic anemia; PRCA, pure red cell aplasia; CAD, cold agglutinin disease; ITP, immune thrombocytopenia; XIAP Def s/p HSCT, X-linked inhibitor of apoptosis protein deficiency status post hematopoietic stem cell transplant; CTLA 4 Def s/p HSCT, cytotoxic T-lymphocyte associated protein 4 deficiency status post hematopoietic stem cell transplant; AIN, autoimmune neutropenia; AIT, autoimmune thrombocytopenia; CGD s/p HSCT, chronic granulomatous disease status post hematopoietic stem cell transplant; Evan’s, Evan’s syndrome; CIDP/SLONM, chronic inflammatory demyelinating polyneuropathy/sporadic late-onset nemaline myopathy; MG, myasthenia gravis; IV, intravenous; CR, complete response; PR, partial response; NR, no response.

Response metrics for the 12 diagnostic groups mirrored that of the overall cohort (Table 6). Complete and partial responses were reported in 53/92 (58%) and 28/92 (30%) of cases, respectively, for a total response rate of 81/92 (88%). Median time to response with daratumumab was 28 days (IQR, 14 to 61 days), and responses were durable (median 270 days; IQR, 123 to 365 days). Mild infusion reactions occurred in 11 cases (11%) (Table 5). No severe allergic reactions or deaths due to daratumumab were reported.

Table 6

Survey of daratumumab use in non-neoplastic immune conditions (n=92)*

Characteristic ITP (n=7) AIHA (n=22) Post Tx AIC (n=9) Post Tx PRCA (n=11) SLE (n=8) ABMR (n=5) CAD (n=4) TTP (n=9) Hem A (n=3) Acquired hemophilia A (n=4) Encephalitis (n=6) M/M/M (n=4)
Prior lines of therapy 4 (3 to 6) 5 (2 to 6) 4 (3 to 5) 2 (0 to 3) 4 (4 to 6) 3 (2 to 3) 5 (3 to 6) 3 (3 to 4) 4 (3 to 4) 4 (3 to 5) 4 (4 to 5) 6 (5 to 6)
Time from diagnosis to daratumumab start (days) 149 (133 to 1,020) 265 (118 to 559) 19 (8 to 56) 270 (184 to 390) 3,285 (2,920 to 3,650) 17 (11 to 24) 2,135 (1,050 to 2,920) 16 (5 to 105) 32 (22 to 32) 413 (301 to 452) 173 (83 to 207) 263 (212 to 536)
Daratumumab doses administered 7 (5 to 8) 4 (3 to 6) 4 (4 to 6) 4 (2 to 5) 22 (13 to 22) 7 (6 to 9) 19 2 (1 to 4) 6 (6 to 7) 16 (13 to 19) 8 (7 to 10) 17 (11 to 20)
Time to daratumumab response (days) 49 (42 to 60) 28 (7 to 60) 18 (8 to 35) 14 (12 to 28) 90 (87 to 90) 56 (28 to 90) 39 (25 to 52) 19 (11 to 27) 12 (10 to 18) 39 (25 to 72) 49 (26 to 59) 44 (26 to 79)
Response§
   Complete 4 [57] 10 [46] 6 [67] 10 [91] 4 [50] 3 [60] 1 [25] 7 [78] 0 4 [100] 2 [33] 2 [50]
   Partial 8 [36] 1 [11] 1 [9] 3 [38] 2 [40] 3 [75] 1 [11] 3 [100] 0 4 [67] 2 [50]
Durability of response (days) 365 (119 to 365) 270 (103 to 412) 187 (100 to 374) 225 (120 to 323) 365 (365 to 678) 203 (140 to 360) 270 and 540 156 (112 to 197) 344 (266 to 367) 186 (137 to 215) 390 (278 to 435) 288 (203 to 386)
Recurrence of primary disorder in follow up 1 [14] 6 [27] 0 0 0 1 [20] 0 2 [22] 2 [67] 0 0 0

Data are presented as n (%) or median (interquartile range). *, only diagnoses for which ≥3 published cases were available are included in this table. All other diagnoses are described in the manuscript. , two antibody-mediated heart and three antibody-mediated renal transplant rejection cases included. , doses of daratumumab were only reported for one patient. §, definitions for response criteria were based on national or international society guidelines, consensus criteria, disease-specific clinical trial definitions, or authors’ discretion when the former were unavailable. , durability of response was only available for two patients. Post Tx, post transplant; AIHA, autoimmune hemolytic anemia; PRCA, pure red cell aplasia; AIC, autoimmune cytopenia; TTP, thrombotic thrombocytopenic purpura; SLE, systemic lupus erythematosus; ITP, immune thrombocytopenia; RBC, red blood cell; Hem A, hemophilia A with inhibitors; M/M/M, myositis/myopathy/myasthenia; CAD, cold agglutinin disease; ABMR, antibody mediated rejection.

Novel use of daratumumab as frontline therapy was reported in one series. Xie et al. described 5 patients with TTP who received daratumumab, corticosteroids, and TPE. Four out of 5 also received rituximab (59). Four of 5 patients experienced an initial clinical response with normalization of their platelet count prior to hospital discharge. Three patients achieved clinical remission. One patient relapsed, and one patient experienced an exacerbation. However, the latter two patients developed clinical remission after additional daratumumab, corticosteroids, and TPE. Of note, these patients were not part of a clinical trial and were treated in a non-US healthcare system.


Conclusions

We conducted a literature review of published cases describing off-label use of daratumumab for the treatment of antibody-mediated, non-neoplastic immune disorders. Most patients received daratumumab after they were deemed refractory to conventional therapy and had received 4 prior lines of therapy. Addition of daratumumab was associated with a significant overall response rate of 89% (58% CR and 31% PR). Overall, time to response was rapid—median 32 days, and durability of response prolonged—median of 270 days. Response durability was likely longer in most cases but limited by duration of follow up. Recurrence rates and immediate infusion reaction rates were low at 14% and 12%, respectively. Infusion reactions were mild, and, in most cases, subsequent doses were administered uneventfully.

CD38+ plasma cells may play a significant role in refractory antibody-mediated disorders. Specifically, LLPCs may be the dominant antibody-producing cells in patients who have been treated with anti-CD20 agents. Mahévas et al. [2013] demonstrated that LLPCs populated splenic tissues in 10 individuals with refractory ITP who had received anti-CD20 therapy. Notably, splenic tissue from 5 individuals who had not received anti-CD20 therapy but who experienced ITP for a similar duration showed mostly short-lived CD19+ plasmablasts (76). Analogous splenic studies performed in individuals with refractory warm AIHA despite anti-CD20 agents also demonstrated increased LLPCs (77). Taken together these studies suggest that administration of anti-CD20 therapy may provide selective pressure to shift the splenic antibody-producing cell milieu towards LLPCs. In these settings, daratumumab may be a reasonable second line therapy.

However, autoreactive plasma cells may persist in locations outside of the spleen which are difficult to access with traditional immunosuppression (78,79). Immature plasma cells (plasmablasts and short-lived plasma cells) which develop in peripheral germinal centers can migrate to the bone marrow, where they are thought to compete with pre-existing LLPCs for survival niches (80), although these processes are poorly understood (23). LLPCs in the bone marrow, which may exhibit greater longevity than LLPCs that develop in the spleen or other inflamed tissues, may contribute to refractoriness in autoimmune disorders (21-23). As immature plasma cells evolve to LLPCs, intermediate forms express CD38 and would likely be susceptible to daratumumab. Daratumumab-mediated plasma cell clearance occurs successfully in the bone marrow as demonstrated by clinical trial of its use in multiple myeloma (81). As noted in this work, most individuals with refractory autoimmune disorders responded to daratumumab, which suggests that these processes are driven CD38+ plasma cells and that daratumumab mediates plasma cell clearance in all niches.

Interestingly, daratumumab also demonstrated efficacy in conditions driven by alloimmunization, which is not a pathologic process per se but an expected immune response following exposure to foreign antigens. The effect of daratumumab on normal plasma cells is unclear and is difficult to evaluate in human subjects, who have received daratumumab for neoplastic processes. Frerichs et al. [2020] demonstrated a significant reduction in normal (non-clonal) plasma cell quantity from baseline to 3 months after initiation of daratumumab and at time of progression in bone marrow biopsy samples from 17 patients with relapsed/refractory multiple myeloma (RRMM) (82). The reduction in normal plasma cells corresponded to reductions in polyclonal IgA, IgM, and IgE levels, but not IgG. The authors speculate that the preserved IgG levels may be the result of a subset of normal plasma cells with reduced CD38 and CD19 expression following daratumumab treatment. Further, daratumumab-treated and naïve patients with RRMM showed similar, suboptimal responses to vaccination, which suggests that despite the inherent immune dysregulation in multiple myeloma, daratumumab does not further degrade the ability to produce normal IgG-producing plasma cells. These observations seem at odds with the findings in this study that daratumumab may be helpful in patients experiencing HLA alloimmunization. We propose several explanations for these observations. HLA antibody producing plasma cells may express CD38 more prominently than other LLPCs in the bone marrow niche and therefore may be more susceptible to daratumumab. Alternatively, HLA antibody-producing plasma cells may make up a smaller proportion of total bone marrow plasma cells. Thus, any loss in this population may result in significant decrement in HLA antibody production. Lastly, HLA antibody-producing plasma cells may be more often located in extramedullary locations with survival niches that are less robust than the bone marrow and therefore are more susceptible to clearance efforts.

Despite the lack of mechanistic understanding of daratumumab activity against normal plasma cells, experience with off-label use of daratumumab is expanding in the transplant community. Antibody-mediated phenomena can impede several steps of the transplant process for hematopoietic stem cell (HSC) and solid organ transplant: (I) ABO isohemagglutinins may cause delayed red cell engraftment in HSC; and solid organ rejection in ABO incompatible transplants; (II) HLA sensitization can narrow the potential donor pool, cause platelet refractoriness, and result in HSC or solid organ transplant rejection; and (III) minor red cell antigen alloimmunization may limit red cell availability for transfusion support. Many of these indications were the subject of published cases included in this review. In post-HSC transplant AICs, post-ABO incompatible HSC transplant PRCA, and HLA antibody-mediated solid organ transplant rejection, daratumumab administration resulted in an overall reported response rate of 92%. We may be able to rationalize these findings in the context of the discussion above. ABO isohemagglutinins are predominantly of IgM isotype, and as described above, IgM isotype normal plasma cells may be uniquely sensitive to daratumumab. In contrast, HLA antibodies are often of IgG isotype and would be predicted to be more difficult to eradicate. However, post-transplant HLA sensitization requires evolution of B cells to immature plasma cells and eventually to antigen-specific LLPCs. There may be many opportunities along this continuum for B- and plasma cell removal prior to establishment of the plasma cell in a durable survival niche. In these patients, daratumumab may be an effective addition to a multimodal anti-humoral rejection strategy.

Fewer cases described use of daratumumab in the pre-transplant setting. Desensitization may be difficult to achieve pre-transplant if HLA or RBC alloantibody production arises from LLPCs in durable survival niches, even with daratumumab use. Only one report detailed success with this approach. Pereda et al. described a case of HLA-immune platelet refractoriness in a patient with aplastic anemia who, after receiving daratumumab, could be supported with platelet transfusions and became eligible for haploidentical transplant (72). The same series described successful daratumumab use to reduce red cell alloimmunization in patients with sickle cell disease prior to allogenic HSC transplant (72). Notably, clinical trials in the United States are recruiting for pre-transplant use of daratumumab in HLA-sensitized renal transplant candidates (NCT04827979) and in sickle cell patients with red cell alloimmunization prior to stem cell transplant (NCT06358638). Data from these clinical trials, continued outcomes reporting, and pre-clinical animal models and in vitro studies will enhance our understanding of the effect of daratumumab on IgG isotype LLPCs and the utility of daratumumab in desensitization regimens.

Our review also identified several cases where daratumumab administration did not improve the underlying antibody-mediated condition or the underlying condition recurred despite initial response. In most of these cases, providers employed traditional B-cell depletion with rituximab prior to administration of daratumumab. Interval CD20+ B-cell quantitation to confirm depletion is not routinely reported; therefore, it is possible that initial “resistance” to daratumumab was due to incomplete CD20+ B-cell depletion or B-cell resistance to anti-CD20 therapy (20). Mechanisms of true resistance to daratumumab, where daratumumab fails to reduce antibody production from plasma cells or when initial response to daratumumab is followed by failure to respond on disease recurrence, in non-malignant conditions are unknown. In patients with multiple myeloma, downregulation of CD38 expression, upregulation of complement inhibitor proteins, and Fcγ receptor mutations play a role in daratumumab resistance (83-86). It is possible that these mechanisms, especially downregulation of CD38 expression, are occurring in normal, healthy or pathologic, autoreactive plasma cells that are exposed to daratumumab. Additional investigation of patients who are initially or subsequently refractory to daratumumab with flow cytometry from bone marrow samples (if available), plasma-drug concentration levels, and drug antibody studies may clarify mechanisms of daratumumab resistance. Understanding these mechanisms may better inform selection of patients who may most benefit from daratumumab and timing of daratumumab administration in the disease course.

Our study had several weaknesses. These include low sample size—particularly for individual disease groups—and the risk of publication bias. Generalization of these results to future cases should occur cautiously. Another weakness is adjudication of response. Autoimmune disorders may have resulted in significant end-organ damage by the time daratumumab was initiated. It therefore becomes difficult to assess clinical response. For example, recovery in the autoimmune myositis/myopathy cases were judged by improvement in strength as measured by an objective scale but were necessarily impacted by severe baseline impairment. Response criteria characterized by antibody detection only, may not capture clinically meaningful responses. In one instance of anti-NMDA-R mediated encephalitis—the clinical response persisted despite gradual re-emergence of antibodies (71). Additionally, standardized response definitions were not always available at the time of publication of each case. The authors sought to standardize response reporting through application of society recommended criteria or consensus definitions where available. For conditions without these the authors applied commonly used response criteria for clinical trials or best judgement to adjudicate responses. For cases which did not present all details included in the response criteria, the authors adjudicated responses with the information available. These adjudications would be expected to have the greatest impact on disease categories with the fewest included cases or where most cases came from the same publication. Lastly, not all included studies occurred in the United States, where insurers can dictate which patients are eligible for off-label therapies. Again, this could impact generalizability of these results to American providers.

In this literature review, we found that daratumumab is a promising therapy for the management of a wide variety of antibody mediated, non-neoplastic immune disorders. Daratumumab mediates removal of antibody-producing plasma cells, which may be responsible for pathologic antibody production in refractory immune conditions. Several case reports and series have been published demonstrating successful use of daratumumab for autoimmune and alloimmune disorders after multiple lines of conventional immunosuppression and anti-CD20 monoclonal therapy and with minimal infusion related-adverse effects. However, publication bias remains a concern. Building the evidence base for daratumumab use in these conditions will be essential to expand the labelled indications for this therapy and ultimately increase its availability for patients with persistent disease despite usual treatment. We encourage high-quality prospective study of daratumumab use in these settings. Baring these, we recommend continued reporting on daratumumab use, performance, and safety as these factors will inform its integration into treatment algorithms and guidelines for antibody-mediated, non-neoplastic immune disorders.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://aob.amegroups.com/article/view/10.21037/aob-24-23/rc

Peer Review File: Available at https://aob.amegroups.com/article/view/10.21037/aob-24-23/prf

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://aob.amegroups.com/article/view/10.21037/aob-24-23/coif). M.H.T. serves as an unpaid editorial board member of Annals of Blood from May 2024 to April 2026. M.H.T. also received speaking and consulting fees from Sanofi for speaking engagements. The other author has no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Davidson A, Diamond B. Autoimmune diseases. N Engl J Med 2001;345:340-50. [Crossref] [PubMed]
  2. Aringer M, Costenbader K, Daikh D, et al. 2019 European League Against Rheumatism/American College of Rheumatology Classification Criteria for Systemic Lupus Erythematosus. Arthritis Rheumatol 2019;71:1400-12. [Crossref] [PubMed]
  3. Narayanaswami P, Sanders DB, Wolfe G, et al. International Consensus Guidance for Management of Myasthenia Gravis: 2020 Update. Neurology 2021;96:114-22. [Crossref] [PubMed]
  4. Garratty G. Mechanisms of immune red cell destruction, and red cell compatibility testing. Hum Pathol 1983;14:204-12. [Crossref] [PubMed]
  5. Thorsby E, Pfeffer P, Leivestad T. Role of HLA molecules in the induction of alloimmune responses: clinical significance in the cyclosporine era. Transplant Proc 2004;36:16S-21S. [Crossref] [PubMed]
  6. Neunert C, Terrell DR, Arnold DM, et al. American Society of Hematology 2019 guidelines for immune thrombocytopenia. Blood Adv 2019;3:3829-66. [Crossref] [PubMed]
  7. Zheng XL, Vesely SK, Cataland SR, et al. ISTH guidelines for treatment of thrombotic thrombocytopenic purpura. J Thromb Haemost 2020;18:2496-502. [Crossref] [PubMed]
  8. Schinstock CA, Mannon RB, Budde K, et al. Recommended Treatment for Antibody-mediated Rejection After Kidney Transplantation: The 2019 Expert Consensus From the Transplantion Society Working Group. Transplantation 2020;104:911-22. [Crossref] [PubMed]
  9. Velleca A, Shullo MA, Dhital K, et al. The International Society for Heart and Lung Transplantation (ISHLT) guidelines for the care of heart transplant recipients. J Heart Lung Transplant 2023;42:e1-e141. [Crossref] [PubMed]
  10. Jäger U, Barcellini W, Broome CM, et al. Diagnosis and treatment of autoimmune hemolytic anemia in adults: Recommendations from the First International Consensus Meeting. Blood Rev 2020;41:100648. [Crossref] [PubMed]
  11. Liu W, Liu J, Xue F, et al. Anti-CD38 antibody for refractory acquired hemophilia A. J Thromb Haemost 2023;21:1515-8. [Crossref] [PubMed]
  12. Syeda MZ, Hong T, Huang C, et al. B cell memory: from generation to reactivation: a multipronged defense wall against pathogens. Cell Death Discov 2024;10:117. [Crossref] [PubMed]
  13. Lobbes H, Lega JC, Le Guenno G, et al. Treatment strategy for acquired pure red cell aplasia: a systematic review and meta-analysis. Blood Adv 2023;7:6451-65. [Crossref] [PubMed]
  14. Cuker A, Cataland SR, Coppo P, et al. Redefining outcomes in immune TTP: an international working group consensus report. Blood 2021;137:1855-61. [Crossref] [PubMed]
  15. Ng JY, Fernando S, Phipps L, et al. Therapeutic Apheresis in Rural and Regional Settings: A Single-Centre 5-Year Experience. Blood 2023;142:3693. [Crossref]
  16. Cavallaro F, Barcellini W, Fattizzo B. Antibody based therapeutics for autoimmune hemolytic anemia. Expert Opin Biol Ther 2023;23:1227-37. [Crossref] [PubMed]
  17. Rodziewicz M, Mendoza-Pinto C, Dyball S, et al. Predictors and prognostic factors influencing outcomes of anti-CD20 monoclonal antibodies in systemic lupus erythematosus: A systematic review update. Semin Arthritis Rheum 2024;65:152346. [Crossref] [PubMed]
  18. Gelbenegger G, Berentsen S, Jilma B. Monoclonal antibodies for treatment of cold agglutinin disease. Expert Opin Biol Ther 2023;23:395-406. [Crossref] [PubMed]
  19. Weiner GJ. Rituximab: mechanism of action. Semin Hematol 2010;47:115-23. [Crossref] [PubMed]
  20. Freeman CL, Sehn LH. A tale of two antibodies: obinutuzumab versus rituximab. Br J Haematol 2018;182:29-45. [Crossref] [PubMed]
  21. Mumtaz IM, Hoyer BF, Panne D, et al. Bone marrow of NZB/W mice is the major site for plasma cells resistant to dexamethasone and cyclophosphamide: implications for the treatment of autoimmunity. J Autoimmun 2012;39:180-8. [Crossref] [PubMed]
  22. Kuwana M, Iki S, Urabe A. The role of autoantibody-producing plasma cells in immune thrombocytopenic purpura refractory to rituximab. Am J Hematol 2007;82:846-8. [Crossref] [PubMed]
  23. Mei HE, Wirries I, Frölich D, et al. A unique population of IgG-expressing plasma cells lacking CD19 is enriched in human bone marrow. Blood 2015;125:1739-48. [Crossref] [PubMed]
  24. Fooksman DR, Jing Z, Park R. New insights into the ontogeny, diversity, maturation and survival of long-lived plasma cells. Nat Rev Immunol 2024;24:461-70. [Crossref] [PubMed]
  25. Hill E, Morrison C, Kazandjian D. Daratumumab: A review of current indications and future directions. Semin Oncol 2022;49:48-59. [Crossref] [PubMed]
  26. Research C for DE and. Daratumumab (DARZALEX). FDA [Internet]. 2019 Feb 9 [cited 2024 Jul 7]. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/daratumumab-darzalex
  27. Dossier C, Prim B, Moreau C, et al. A global antiB cell strategy combining obinutuzumab and daratumumab in severe pediatric nephrotic syndrome. Pediatr Nephrol 2021;36:1175-82. [Crossref] [PubMed]
  28. Alexander T, Ostendorf L, Biesen R, et al. Sustained responses after anti-CD38 treatment with daratumumab in two patients with refractory systemic lupus erythematosus. Ann Rheum Dis 2023;82:1497-9. [Crossref] [PubMed]
  29. Scheibe F, Ostendorf L, Reincke SM, et al. Daratumumab treatment for therapy-refractory anti-CASPR2 encephalitis. J Neurol 2020;267:317-23. [Crossref] [PubMed]
  30. Koo J, Giller RH, Quinones R, et al. Autoimmune cytopenias following allogeneic hematopoietic stem cell transplant in pediatric patients: Response to therapy and late effects. Pediatr Blood Cancer 2020;67:e28591. [Crossref] [PubMed]
  31. Malkiel S, Wheaton L, Calderwood L, et al. Daratumumab Effectively Treats Refractory Immune Hemolytic Anemia after Hematopoietic Stem Cell Transplantation: Case Report. [cited 2024 Jul 7]. Available online: https://austinpublishinggroup.com/hematology/fulltext/hematology-v7-id1292.php
  32. Rieger MJ, Stolz SM, Ludwig S, et al. Daratumumab in rituximab-refractory autoimmune haemolytic anaemia. Br J Haematol 2021;194:931-4. [Crossref] [PubMed]
  33. Tolbert VP, Goldsby R, Huang J, et al. Daratumumab Is Effective in the Treatment of Refractory Post-Transplant Autoimmune Hemolytic Anemia: A Pediatric Case Report. Blood 2016;128:4819. [Crossref]
  34. Blennerhassett R, Sudini L, Gottlieb D, et al. Post-allogeneic transplant Evans syndrome successfully treated with daratumumab. Br J Haematol 2019;187:e48-51. [Crossref] [PubMed]
  35. Schuetz C, Hoenig M, Moshous D, et al. Daratumumab in life-threatening autoimmune hemolytic anemia following hematopoietic stem cell transplantation. Blood Adv 2018;2:2550-3. [Crossref] [PubMed]
  36. Driouk L, Schmitt R, Peters A, et al. Daratumumab therapy for post-HSCT immune-mediated cytopenia: experiences from two pediatric cases and review of literature. Mol Cell Pediatr 2021;8:5. [Crossref] [PubMed]
  37. Khandelwal P, Teusink-Cross A, Kumar AR, et al. Daratumumab for the management of autoimmune cytopenias in children and young adults: a case series. Br J Haematol 2021;194:e84-9. [Crossref] [PubMed]
  38. Cooling L, Hugan S. Daratumumab in combination with standard treatment for autoimmune hemolytic anemia in a pediatric patient. Transfusion 2019;59:3801-2. [Crossref] [PubMed]
  39. Jain A, Gupta DK. Daratumumab for refractory warm autoimmune hemolytic anemia. Ann Hematol 2021;100:1351-3. [Crossref] [PubMed]
  40. Even-Or E, Naser Eddin A, Shadur B, et al. Successful treatment with daratumumab for post-HSCT refractory hemolytic anemia. Pediatr Blood Cancer 2020;67:e28010. [Crossref] [PubMed]
  41. Migdady Y, Ediriwickrema A, Jackson RP, et al. Successful treatment of thrombocytopenia with daratumumab after allogeneic transplant: a case report and literature review. Blood Adv 2020;4:815-8. [Crossref] [PubMed]
  42. Landon-Cardinal O, Allard-Chamard H, Chapdelaine H, et al. Daratumumab as a rescue therapy in severe refractory anti-SRP immune-mediated necrotising myopathy. Ann Rheum Dis 2023;82:579-80. [Crossref] [PubMed]
  43. Holzer MT, Nies JF, Oqueka T, et al. Successful Rescue Therapy With Daratumumab in Rapidly Progressive Interstitial Lung Disease Caused by MDA5-Positive Dermatomyositis. Chest 2023;163:e1-5. [Crossref] [PubMed]
  44. Scheibe F, Ostendorf L, Prüss H, et al. Daratumumab for treatment-refractory antibody-mediated diseases in neurology. Eur J Neurol 2022;29:1847-54. [Crossref] [PubMed]
  45. Vernava I, Schmitt CA. Daratumumab as a novel treatment option in refractory ITP. Blood Cells Mol Dis 2023;99:102724. [Crossref] [PubMed]
  46. Canales-Herrerias P, Crickx E, Broketa M, et al. High-affinity autoreactive plasma cells disseminate through multiple organs in patients with immune thrombocytopenic purpura. J Clin Invest 2022;132:e153580. [Crossref] [PubMed]
  47. Strüßmann T, Jung J, Heinz J, et al. Long-term complete remission of refractory severe idiopathic immune thrombocytopenia (ITP) treated with daratumumab. Ann Hematol 2023;102:245-7. [Crossref] [PubMed]
  48. Tomkins O, Berentsen S, Arulogun S, et al. Daratumumab for disabling cold agglutinin disease refractory to B-cell directed therapy. Am J Hematol 2020;95:E293-5. [Crossref] [PubMed]
  49. Mohamed A, Alkhatib M, Alshurafa A, et al. Refractory cold agglutinin disease successfully treated with daratumumab. A case report and review of literature. Hematology 2023;28:2252651. [Crossref] [PubMed]
  50. Shen KN, Zhao DQ, Mao YY, et al. Daratumumab-based regimen for cold agglutinin disease refractory to therapy directing B-cell clone. Leuk Lymphoma 2024;65:410-2. [Crossref] [PubMed]
  51. Zaninoni A, Giannotta JA, Gallì A, et al. The Immunomodulatory Effect and Clinical Efficacy of Daratumumab in a Patient With Cold Agglutinin Disease. Front Immunol 2021;12:649441. [Crossref] [PubMed]
  52. Ostendorf L, Burns M, Durek P, et al. Targeting CD38 with Daratumumab in Refractory Systemic Lupus Erythematosus. N Engl J Med 2020;383:1149-55. [Crossref] [PubMed]
  53. Roccatello D, Fenoglio R, Caniggia I, et al. Daratumumab monotherapy for refractory lupus nephritis. Nat Med 2023;29:2041-7. [Crossref] [PubMed]
  54. Doberer K, Kläger J, Gualdoni GA, et al. CD38 Antibody Daratumumab for the Treatment of Chronic Active Antibody-mediated Kidney Allograft Rejection. Transplantation 2021;105:451-7. [Crossref] [PubMed]
  55. Kwun J, Matignon M, Manook M, et al. Daratumumab in Sensitized Kidney Transplantation: Potentials and Limitations of Experimental and Clinical Use. J Am Soc Nephrol 2019;30:1206-19. [Crossref] [PubMed]
  56. Süsal CC, Kraft L, Ender A, et al. Blood group-specific apheresis in combination with daratumumab as a rescue therapy of acute antibody-mediated rejection in a case of ABO- and human leukocyte antigen-incompatible kidney transplantation. SAGE Open Med Case Rep 2023;11:2050313X231211050.
  57. Aggarwal A, White D, Pavord S, et al. Daratumumab for refractory immune-mediated thrombotic thrombocytopenic purpura. Br J Haematol 2023;202:429-33. [Crossref] [PubMed]
  58. van den Berg J, Kremer Hovinga JA, Pfleger C, et al. Daratumumab for immune thrombotic thrombocytopenic purpura. Blood Adv 2022;6:993-7. [Crossref] [PubMed]
  59. Xie XT, Xiao YY, Zhang Y, et al. Combination regimens containing daratumumab for initial diagnosed acquired thrombotic thrombocytopenic purpura. J Thromb Thrombolysis 2023;55:399-405. [Crossref] [PubMed]
  60. Moonla C, Uaprasert N, Watanaboonyongcharoen P, et al. Daratumumab rapidly reduces high-titre factor VIII inhibitors in haemophilia A patients during life-threatening haemorrhages. Haemophilia 2021;27:e155-9. [Crossref] [PubMed]
  61. Yates B, Molloy E, Dulau-Florea A, et al. Daratumumab for delayed RBC engraftment following major ABO mismatched haploidentical bone marrow transplantation. Transfusion 2021;61:1041-6. [Crossref] [PubMed]
  62. Salas MQ, Alahmari A, Lipton JH. Successful treatment of refractory red cell aplasia after allogeneic hematopoietic cell transplantation with daratumumab. Eur J Haematol 2020;104:145-7. [Crossref] [PubMed]
  63. Chapuy CI, Kaufman RM, Alyea EP, et al. Daratumumab for Delayed Red-Cell Engraftment after Allogeneic Transplantation. N Engl J Med 2018;379:1846-50. [Crossref] [PubMed]
  64. Asawapanumas T, Chanswangphuwana C, Watanaboonyongcharoen P, et al. Daratumumab as a Frontline Immunosuppression for Pure Red Cell Aplasia after Major ABO-mismatched Allogeneic Hematopoietic Stem Cell Transplantation. Leuk Res Rep 2022;17:100314. [Crossref] [PubMed]
  65. Henig I, Yehudai-Ofir D, Zohar Y, et al. Pure Red Cell Aplasia following ABO-Mismatched Allogeneic Hematopoietic Stem Cell Transplantation: Resolution with Daratumumab Treatment. Acta Haematol 2021;144:683-7. [Crossref] [PubMed]
  66. Rautenberg C, Kaivers J, Germing U, et al. Daratumumab for treatment of pure red cell aplasia after allogeneic stem cell transplantation. Bone Marrow Transplant 2020;55:1191-3. [Crossref] [PubMed]
  67. Jeyaraman P, Borah P, Rajput P, et al. Daratumumab for pure red cell aplasia post ABO incompatible allogeneic hematopoietic stem cell transplant for aplastic anemia. Blood Cells Mol Dis 2021;88:102464. [Crossref] [PubMed]
  68. Martino R, García-Cadenas I, Esquirol A. Daratumumab may be the most effective treatment for post-engraftment pure red cell aplasia due to persistent anti-donor isohemagglutinins after major ABO-mismatched allogeneic transplantation. Bone Marrow Transplant 2022;57:282-5. [Crossref] [PubMed]
  69. Bathini S, Holtzman NG, Koka R, et al. Refractory postallogeneic stem cell transplant pure red cell aplasia in remission after treatment with daratumumab. Am J Hematol 2019;94:E216-9. [Crossref] [PubMed]
  70. Bicsko RR, Magyari F, Szasz R, et al. Administration of Daratumumab in the Case of Severe Pure Red Cell Aplasia after Allogeneic Transplantation. [cited 2024 Jul 7]. Available online: https://austinpublishinggroup.com/hematology/fulltext/hematology-v8-id1331.php
  71. Ratuszny D, Skripuletz T, Wegner F, et al. Case Report: Daratumumab in a Patient With Severe Refractory Anti-NMDA Receptor Encephalitis. Front Neurol 2020;11:602102. [Crossref] [PubMed]
  72. Pereda MA, Hosahalli Vasanna S, Desai NJ, et al. Case report: Daratumumab treatment in pre-transplant alloimmunization and severe hemolytic anemia. Front Immunol 2022;13:1055473. [Crossref] [PubMed]
  73. Epperly R, Santiago T, Morin CE, et al. Targeting plasma cells with daratumumab aids in the treatment of post-transplant autoimmune-like hepatitis. Pediatr Blood Cancer 2021;68:e29290. [Crossref] [PubMed]
  74. Pleguezuelo DE, Díaz-Simón R, Cabrera-Marante O, et al. Case Report: Resetting the Humoral Immune Response by Targeting Plasma Cells With Daratumumab in Anti-Phospholipid Syndrome. Front Immunol 2021;12:667515. [Crossref] [PubMed]
  75. Mathew Thomas V, Gilreath JA, Newton S, et al. Daratumumab as a novel treatment for refractory acquired von Willebrand syndrome associated with monoclonal gammopathy. Haemophilia 2021;27:e563-6. [Crossref] [PubMed]
  76. Mahévas M, Patin P, Huetz F, et al. B cell depletion in immune thrombocytopenia reveals splenic long-lived plasma cells. J Clin Invest 2013;123:432-42. [Crossref] [PubMed]
  77. Mahévas M, Michel M, Vingert B, et al. Emergence of long-lived autoreactive plasma cells in the spleen of primary warm auto-immune hemolytic anemia patients treated with rituximab. J Autoimmun 2015;62:22-30. [Crossref] [PubMed]
  78. Khodadadi L, Cheng Q, Alexander T, et al. Bortezomib Plus Continuous B Cell Depletion Results in Sustained Plasma Cell Depletion and Amelioration of Lupus Nephritis in NZB/W F1 Mice. PLoS One 2015;10:e0135081. [Crossref] [PubMed]
  79. Taddeo A, Khodadadi L, Voigt C, et al. Long-lived plasma cells are early and constantly generated in New Zealand Black/New Zealand White F1 mice and their therapeutic depletion requires a combined targeting of autoreactive plasma cells and their precursors. Arthritis Res Ther 2015;17:39. [Crossref] [PubMed]
  80. Chernova I, Jones DD, Wilmore JR, et al. Lasting antibody responses are mediated by a combination of newly formed and established bone marrow plasma cells drawn from clonally distinct precursors. J Immunol 2014;193:4971-9. [Crossref] [PubMed]
  81. Lokhorst HM, Plesner T, Laubach JP, et al. Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma. N Engl J Med 2015;373:1207-19. [Crossref] [PubMed]
  82. Frerichs KA, Bosman PWC, van Velzen JF, et al. Effect of daratumumab on normal plasma cells, polyclonal immunoglobulin levels, and vaccination responses in extensively pre-treated multiple myeloma patients. Haematologica 2020;105:e302-6. [Crossref] [PubMed]
  83. Perez de Acha O, Reiman L, Jayabalan DS, et al. CD38 antibody re-treatment in daratumumab-refractory multiple myeloma after time on other therapies. Blood Adv 2023;7:6430-40. [Crossref] [PubMed]
  84. Nijhof IS, Casneuf T, van Velzen J, et al. CD38 expression and complement inhibitors affect response and resistance to daratumumab therapy in myeloma. Blood 2016;128:959-70. [Crossref] [PubMed]
  85. Franssen LE, Stege CAM, Zweegman S, et al. Resistance Mechanisms Towards CD38-Directed Antibody Therapy in Multiple Myeloma. J Clin Med 2020;9:1195. [Crossref] [PubMed]
  86. Ghose J, Viola D, Terrazas C, et al. Daratumumab induces CD38 internalization and impairs myeloma cell adhesion. Oncoimmunology 2018;7:e1486948. [Crossref] [PubMed]
doi: 10.21037/aob-24-23
Cite this article as: Jones JM, Tran MH. A literature review of the use of daratumumab in non-neoplastic immunologic disorders. Ann Blood 2024;9:33.

Download Citation