Fact-checked by Grok 2 weeks ago

Daratumumab

Daratumumab is a human IgG1κ monoclonal antibody that specifically binds to , a transmembrane highly expressed on malignant plasma cells in , inducing tumor cell through , antibody-dependent cell-mediated cytotoxicity, antibody-dependent cellular , and direct . It was first approved by the U.S. in November 2015 as monotherapy for patients with who had received at least three prior lines of therapy, marking it as the first CD38-targeted agent in its class. Subsequent approvals expanded its use to frontline and relapsed settings, often in combination with inhibitors, immunomodulatory drugs, or corticosteroids, demonstrating improved and overall response rates in clinical trials such as , POLLUX, and . A subcutaneous , daratumumab and hyaluronidase-fihj (Darzalex Faspro), was approved in for broader accessibility, reducing times while maintaining . Common adverse effects include -related reactions, , , and infections, necessitating premedication and monitoring, though its risk-benefit profile has established it as a cornerstone in therapy.

Medical Applications

Indications and Approvals

Daratumumab received accelerated approval from the (FDA) on November 16, 2015, as monotherapy for adult patients with who have received at least three prior lines of therapy, including a and an immunomodulatory agent, or who are double- to both classes. This was based on response rates in heavily pretreated relapsed or (RRMM) patients. The (EMA) granted conditional marketing authorization for the same monotherapy indication in RRMM on May 20, 2016, for patients with at least three prior therapies. Subsequent FDA expansions included approval on January 31, 2019, for combination with and dexamethasone (D-Rd) in RRMM patients after at least one prior line of therapy. On June 27, 2019, the FDA approved daratumumab with , , and (D-VMP) for newly diagnosed (NDMM) patients ineligible for autologous transplant. Further, on September 26, 2019, approval extended to daratumumab with , , and dexamethasone (D-VTd) for NDMM patients eligible for transplant, and on September 20, 2020, to daratumumab with and dexamethasone for RRMM after at least two prior lines. The EMA aligned with similar combination approvals, including D-Rd for RRMM after one prior line on June 28, 2019, and D-VMP for transplant-ineligible NDMM on December 18, 2018. The subcutaneous formulation, daratumumab and hyaluronidase-fihj (Darzalex Faspro), was FDA-approved on May 1, 2020, initially for monotherapy and later expanded to match intravenous indications, including RRMM combinations after one or more prior lines and NDMM frontline regimens. On July 30, 2024, the FDA approved Darzalex Faspro with , , and dexamethasone (D-VRd) for induction and consolidation in transplant-eligible NDMM patients. EMA approvals for the subcutaneous form followed, including D-VTd for transplant-eligible NDMM on January 27, 2020, and extensions to D-VRd. In 2025, the approved subcutaneous daratumumab monotherapy on July 23 for adults with high-risk smoldering , marking the first licensed treatment for this precursor condition, based on data in asymptomatic patients at high risk of progression. This targets patients without active myeloma symptoms but with biomarkers indicating elevated risk, such as involvement exceeding 60% or free light chain ratio greater than 100. FDA consideration for this indication received Oncologic Drugs Advisory Committee support in June 2025 but remained pending full approval as of October 2025.

Clinical Efficacy Evidence

In the phase 3 POLLUX trial evaluating daratumumab plus and dexamethasone (D-Rd) versus and dexamethasone (Rd) in patients with relapsed or refractory (RRMM) who had received at least one prior therapy, the overall response rate (ORR) was 93% with D-Rd compared to 76% with Rd, including higher rates of very good partial response or better (81% vs. 49%). D-Rd reduced the risk of progression or death by 63%, with a (HR) for (PFS) of 0.37 after a median follow-up of 13.5 months, and longer-term data confirmed sustained PFS benefits. The phase 3 trial assessed daratumumab plus and dexamethasone (D-Vd) versus and dexamethasone (Vd) in RRMM patients with one to three prior lines of therapy, yielding an ORR of 83% with D-Vd versus 63% with Vd, and PFS of 18.0 months versus 7.3 months in that . Extended follow-up at 42 months showed PFS rates of 22% with D-Vd versus 1% with Vd, alongside deeper responses including higher (MRD) negativity rates. For newly diagnosed (NDMM) in transplant-eligible patients, the phase 3 trial demonstrated that daratumumab plus , , and dexamethasone (D-VRd) induction followed by daratumumab plus (DR) maintenance improved PFS compared to VRd alone, with PFS projected at 17 years in the D-VRd arm based on 2025 projections and sustained MRD negativity in nearly two-thirds of patients achieving over 95% 48-month PFS rates. At a follow-up of 47.5 months, D-VRd yielded higher rates of MRD negativity at 10^{-5} and 10^{-6} thresholds. Subgroup analyses from trials like PERSEUS indicate daratumumab-containing regimens provide PFS benefits in patients with high-risk cytogenetic abnormalities (HRCAs, including del(17p), t(4;14), t(14;16), or gain(1q)), though outcomes may be attenuated compared to standard-risk patients; for instance, 1q gain or amplification, present in up to 40% of cases, correlates with reduced CD38 expression and potentially inferior responses despite overall HR reductions. In high-risk smoldering , the phase 3 AQUILA trial of subcutaneous daratumumab monotherapy versus active monitoring reported a 51% lower risk of progression to active myeloma or death after median follow-up exceeding 5 years (65 months), with continued single-agent activity observed at around 7 years but limited long-term overall survival data due to the disease's indolent nature. Real-world studies in RRMM report ORRs of 76-93% with daratumumab-based regimens, varying by prior refractoriness and combination (e.g., 76.3% overall in relapsed settings, lower at 30% for monotherapy), with median PFS ranging from 6-28 months depending on lines of therapy and cytogenetic risk, though these outcomes reflect heterogeneous populations and shorter follow-up than pivotal trials.

Comparative Effectiveness

In relapsed/refractory multiple myeloma (RRMM), daratumumab plus bortezomib and dexamethasone (D-Vd) showed superior progression-free survival (PFS) over Vd alone in the phase 3 CASTOR trial, with a median PFS of 16.6 months versus 7.9 months (hazard ratio [HR] 0.39; 95% CI 0.33-0.47) and an overall survival (OS) benefit (HR 0.74; 95% CI 0.59-0.92 at 3-year follow-up). Similarly, in the POLLUX trial, daratumumab plus lenalidomide and dexamethasone (D-Rd) extended median PFS to 18.4 months compared to 9.3 months with Rd alone (HR 0.44; 95% CI 0.35-0.55), though OS gains were more modest in heavily pretreated subgroups. These head-to-head data indicate daratumumab adds approximately 7-9 months of PFS when combined with proteasome inhibitor or immunomodulatory drug backbones, driven by deeper responses in CD38-expressing disease, but real-world analyses suggest attenuated OS benefits in triple-class refractory settings due to subsequent therapies. Meta-analyses of daratumumab combinations versus -, -, or -based regimens confirm consistent PFS improvements (pooled HR 0.40-0.50 across RRMM trials), with matching-adjusted indirect comparisons showing daratumumab monotherapy or D-Rd outperforming plus low-dose dexamethasone in OS for heavily pretreated patients (HR 0.72). In newly diagnosed ineligible for transplant, D-Rd reduced progression risk versus , , and dexamethasone (VRd) (HR 0.59 for PFS), though direct superiority in OS remains unproven without mature data. Efficacy depends on baseline expression, with reduced responses in low-CD38 tumors, underscoring no universal advantage over backbone therapies in all causal pathways of disease progression. Compared to bispecific antibodies (e.g., ) or CAR-T therapies (e.g., cilta-cel) in daratumumab-refractory RRMM, daratumumab regimens offer earlier-line, outpatient administration with lower toxicity but shallower responses (complete response rates 10-20% lower) and shorter PFS in triple-class exposed patients (median 6-12 months versus 18-24 months for CAR-T). Indirect comparisons favor CAR-T for OS in refractory cases (HR 0.50-0.70 versus daratumumab historical controls), reflecting T-cell redirection's independence from targeting, though daratumumab's role persists as a less resource-intensive bridge to these modalities.
Trial/RegimenComparisonMedian PFS (months)PFS HR (95% CI)OS HR (if available)
(D-Vd vs Vd, RRMM)Post-1L16.6 vs 7.90.39 (0.33-0.47)0.74 (0.59-0.92)
POLLUX (D-Rd vs Rd, RRMM)Post-1L IMiD18.4 vs 9.30.44 (0.35-0.55)Modest gain
APOLLO (D-Vd vs Vd, RRMM)Post-Len/Pom11.2 vs 7.10.63 (0.50-0.79)0.79 (0.60-1.03)

Safety and Tolerability

Common Adverse Effects

The most common adverse effects of daratumumab in patients, observed across pivotal phase 3 trials such as POLLUX (daratumumab plus and dexamethasone), (daratumumab plus and dexamethasone), and (daratumumab plus and dexamethasone), include infusion-related reactions, , , , upper respiratory tract infections, and . These effects are generally manageable with monitoring and supportive care, with hematologic toxicities showing reversibility in the majority of cases following dose interruption or adjustment. Infusion-related reactions with intravenous daratumumab occurred in 41% to 48% of patients overall, with incidences of 45% to 56% specifically at the first dose; most were grade 1 or 2 (e.g., , , chills, or throat irritation), and grade 3 events ranged from 2% to 9%. protocols, including intravenous corticosteroids (e.g., dexamethasone 20 mg or 100 mg), antipyretics (e.g., acetaminophen 650-1000 mg), and antihistamines (e.g., diphenhydramine 25-50 mg), administered 1-3 hours prior to , along with post-infusion oral corticosteroids for the first two cycles, substantially mitigate these reactions; infusion rate reductions or interruptions are recommended for symptomatic management. In contrast, the subcutaneous formulation (daratumumab with hyaluronidase-fihj) yields lower rates of administration-related reactions, at 12.7% to 13% overall, primarily mild and not requiring routine infusion rate adjustments. Hematologic cytopenias are frequent and regimen-dependent: affected 40% to 48% of patients (13% grade 3/4), 50% to 91% (12% to 56% grade 3/4), and 67% to 90% (3% to 47% grade 3/4), with periodic monitoring advised to guide holding or discontinuing therapy if severe. Upper infections occurred in 44% to 52% of patients (2% to 6% grade 3/4), often linked to from cytopenias or concurrent therapies. was reported in 35% to 40% (up to 8% grade 3/4), typically self-limiting but contributing to dose reductions in some cases. Post-marketing data align with trial findings, confirming these as the predominant non-serious events without emerging patterns of increased severity.

Serious Risks and Management

Daratumumab treatment is associated with an elevated risk of serious infections, including and upper infections, occurring in approximately 38% of patients overall, due to CD38-mediated depletion of immune cells leading to and . , often exacerbated by concomitant therapies, increases susceptibility to bacterial infections and , with grade 3/4 reported in up to 40% of cases in combination regimens. Management includes routine monitoring of complete blood counts, (G-CSF) prophylaxis or treatment for severe , and prophylaxis such as acyclovir or valacyclovir for zoster prevention, alongside consideration of antibacterial agents like levofloxacin or trimethoprim-sulfamethoxazole for high-risk patients. Intravenous immunoglobulin (IVIG) supplementation may be employed for profound to mitigate recurrent infections. Hepatitis B virus (HBV) reactivation, sometimes fatal, has been observed in less than 1% of patients, particularly those with resolved infection, necessitating pre-treatment screening for HBV surface , core , and DNA levels, followed by antiviral prophylaxis (e.g., entecavir) and ongoing monitoring in at-risk individuals. is rare but reported in post-marketing surveillance, warranting at baseline and periodically during therapy. Infusion-related reactions (IRRs), manifesting as dyspnea, , or in severe cases (affecting <5% for serious events), arise from cytokine release and complement activation, with higher incidence during initial doses. Premedication with corticosteroids, antihistamines, and antipyretics is standard, alongside gradual infusion rate escalation after the first cycle; for , immediate discontinuation, epinephrine, and supportive care are required, with rechallenge possible after resolution under close supervision. In long-term follow-ups from phase 3 trials reported through 2025, approximately 5-15% of patients discontinued due to serious toxicities, primarily infections or IRRs, underscoring the need for vigilant risk-benefit assessment and dose adjustments or interruptions based on toxicity grading. Secondary malignancies linked to prolonged immunosuppression have been noted anecdotally but lack robust incidence data beyond background risks, with no clear causal elevation attributable solely to daratumumab in controlled studies.

Long-Term Safety Data

In extended follow-up analyses of pivotal trials such as and , the long-term safety profile of daratumumab in multiple myeloma patients has shown no emergence of novel adverse events beyond those observed in initial phases, with cumulative exposure highlighting persistent but plateauing risks associated with immune modulation. In the trial's primary analysis, second primary malignancies occurred in 10.7% of patients receiving daratumumab plus VRd compared to 7.2% in the VRd arm, a pattern consistent in maintenance phases without disproportionate escalation over time. Similarly, the 6-year update reported second primary malignancies in 11.4% of the daratumumab-containing arm during maintenance, attributable in part to prolonged immunosuppression from CD38-targeted depletion of immune effector cells alongside lenalidomide's known oncogenic risks. Sustained cytopenias, including neutropenia and thrombocytopenia, exhibit higher grade 3/4 incidence with daratumumab maintenance (e.g., 54.2% versus 46.9% with lenalidomide alone), but real-world data indicate stabilization without indefinite progression, linked causally to cumulative dosing's impact on hematopoiesis rather than irreversible marrow toxicity. Infection rates, elevated early due to NK cell and B-cell subset depletion, plateau after approximately 6 months yet remain 20-30% higher than comparators, as evidenced by meta-analyses and registry observations through 2025, underscoring chronic vulnerability from impaired humoral immunity without evidence of late-onset surges. Diverging from short-term profiles dominated by infusion-related reactions, long-term data reveal greater persistence of fatigue (reported in up to 40% of extended maintenance recipients), potentially stemming from ongoing metabolic or inflammatory sequelae of , contrasted by diminished acute hypersensitivity over cycles. Real-world registries in 2024-2025, including Korean and European cohorts, corroborate trial findings with comparable durability of known risks and absence of unanticipated signals, though empirical scrutiny warrants heightened vigilance for chronic immunosuppression in elderly patients, where baseline frailty amplifies infection and cytopenia causality independent of trial selection biases.

Interactions and Diagnostic Considerations

Pharmacokinetic Interactions

Daratumumab, a monoclonal antibody, undergoes catabolic degradation rather than cytochrome P450 (CYP450)-mediated metabolism, resulting in negligible inhibition or induction of CYP enzymes and limited potential for CYP450-based pharmacokinetic interactions. Clinical assessments in multiple myeloma regimens confirm no significant pharmacokinetic alterations when combined with , , or , standard agents in polypharmacy for the disease. Although formal drug-drug interaction studies have not been conducted, daratumumab's exposure remains consistent in these combinations, supporting its safe co-administration without dose adjustments for these partners. The subcutaneous formulation of daratumumab, co-administered with recombinant human (rHuPH20), enhances dispersion and absorption through subcutaneous tissue by temporarily degrading , achieving bioavailability comparable to intravenous administration without meaningful changes in overall pharmacokinetic profile or steady-state concentrations. This facilitates faster administration while maintaining equivalent systemic exposure, with no reported pharmacokinetic conflicts arising from the enzyme's activity. Population pharmacokinetic analyses indicate that daratumumab's steady-state levels are unaffected by renal or mild hepatic impairment, conditions prevalent in up to 50% of multiple myeloma patients at baseline, obviating the need for dose modifications in these populations. Age, race, and mild hepatic dysfunction similarly exert no clinically relevant impact on exposure, underscoring daratumumab's pharmacokinetic stability amid common comorbidities. For concomitant medications metabolized via strong CYP3A4 inducers or inhibitors, monitoring is advised due to potential indirect effects on polypharmacy partners, though daratumumab itself poses minimal risk.

Interference with Laboratory Tests

Daratumumab binds to CD38, which is expressed at low levels on the surface of red blood cells (RBCs), leading to interference with indirect antiglobulin testing (IAT, also known as the indirect Coombs test) used for antibody screening and crossmatching in pre-transfusion compatibility assessments. This binding causes a positive direct antiglobulin test (DAT) in approximately 70-80% of treated patients and pan-reactivity in serum antibody screens, potentially masking clinically significant alloantibodies against minor antigens while ABO and RhD typing remain unaffected. To mitigate this interference, RBCs can be pretreated with 0.2 M dithiothreitol (DTT), which denatures and eliminates the pan-reactivity without affecting detection of most clinically relevant antibodies, though it may inactivate certain antigens like . Alternatively, daratumumab-specific immunoadsorption reagents, such as , selectively remove the drug from serum to resolve interference in IAT. Guidelines recommend establishing blood type and screen prior to initiating daratumumab therapy, with validated DTT or reagent-based methods for ongoing transfusion needs; for emergencies, administer ABO/RhD-compatible RBCs without crossmatch if necessary. These approaches, validated internationally since 2016, ensure transfusion safety despite persistent interference lasting up to 6 months post-treatment. In flow cytometry-based minimal residual disease (MRD) assessment for multiple myeloma, daratumumab coats CD38-high plasma cells, causing masking that precludes accurate enumeration and risks false-negative results. Validated strategies include using CD38-independent gating markers (e.g., CD138, CD27), anti-CD38 nanobodies like JK36 for specific detection, or temporarily holding therapy 3-6 months prior to testing to allow drug clearance. Such adaptations maintain MRD sensitivity at 10^{-5} to 10^{-6} levels, as demonstrated in studies adjusting for therapeutic monoclonal antibody persistence.

Pharmacology

Mechanism of Action

Daratumumab is a human immunoglobulin G1κ (IgG1κ) monoclonal antibody that binds with high affinity to a unique epitope on the CD38 transmembrane glycoprotein, which is highly expressed on malignant plasma cells in multiple myeloma. CD38 functions as a multifunctional ectoenzyme involved in calcium signaling and cell adhesion, but its overexpression on myeloma cells—often exceeding 90% of surface coverage—renders them particularly susceptible to antibody-mediated targeting without inherent off-target proliferative inhibition. This binding specificity exploits the ubiquity of CD38 on plasma cells as a causal driver of selective cytotoxicity. The primary mechanisms of action involve Fc-dependent immune effector functions: antibody-dependent cellular cytotoxicity (ADCC) mediated by natural killer cells and macrophages, complement-dependent cytotoxicity (CDC) through activation of the classical complement pathway, and antibody-dependent cellular phagocytosis (ADCP) by monocytes and macrophages. These pathways collectively recruit and activate immune cells to lyse CD38-positive targets, with empirical data showing potency scaling with CD38 density on tumor cells. Additionally, daratumumab induces direct apoptosis in CD38-expressing cells via antibody cross-linking, which disrupts signaling and promotes programmed cell death independent of immune effectors. This multifaceted biochemistry underscores daratumumab's reliance on intact CD38 expression for efficacy, as reduced antigen levels diminish ADCC and CDC responses.

Pharmacokinetics and Administration

Daratumumab exhibits nonlinear pharmacokinetics following intravenous (IV) administration, primarily due to target-mediated drug disposition, with clearance decreasing as a function of dose and time owing to saturation and depletion of antigens on target cells. The mean linear clearance is approximately 171 mL/day, and the volume of distribution is limited to about 4.7 L, indicating primary confinement to the vascular and extracellular spaces. Metabolism follows typical pathways for immunoglobulin G1 kappa monoclonal antibodies, involving proteolytic catabolism into peptides and amino acids without cytochrome P450 involvement or significant renal/hepatic excretion; elimination is dominated by the nonlinear target-mediated component alongside a parallel linear process. The terminal half-life associated with the linear clearance phase is approximately 18 days (standard deviation 9 days), though effective half-life extends to around 20-21 days at steady-state concentrations achieved after roughly 5 months of intermittent dosing. Standard IV dosing is weight-based at 16 mg/kg, administered weekly for 6-9 weeks followed by biweekly for up to 16 weeks and then every 4 weeks, allowing accumulation (ratio ~1.6 for maximum concentration) to reach plateau levels with minimal further empirical buildup after initial cycles. Subcutaneous (SC) daratumumab, formulated as and approved by the FDA in May 2020, uses a fixed dose of 1800 mg co-administered with recombinant human hyaluronidase, yielding an absolute bioavailability of approximately 70% and peak concentrations delayed to 3-4 days post-injection. This route reduces administration time to 3-5 minutes via abdominal injection, versus 3-7 hours for initial IV infusions (shorter for subsequent doses), while achieving comparable trough concentrations and efficacy to IV without requiring body weight adjustments. The SC half-life mirrors IV at ~20 days, with similar nonlinear clearance dynamics and accumulation patterns upon repeated dosing per the same schedule.

Pharmacodynamics

Daratumumab, a human IgG1κ monoclonal antibody targeting , induces pharmacodynamic effects through multiple immune-mediated mechanisms, including antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), antibody-dependent cellular phagocytosis (ADCP), and direct induction of apoptosis in CD38-expressing cells. Administration results in rapid depletion of peripheral CD38-positive cells, such as malignant plasma cells and immunosuppressive regulatory B cells, with this depletion correlating directly with clinical responses in patients. The process involves initial engagement of natural killer (NK) cells for ADCC against target cells, though NK cell counts subsequently decline due to their own CD38 expression, highlighting a dynamic balance in immune effector function. Biomarker studies from clinical trial subsets demonstrate that higher baseline CD38 expression density on myeloma cells predicts deeper responses to daratumumab, with patients achieving partial response or better exhibiting significantly elevated pretreatment CD38 levels compared to non-responders. This correlation underscores the causal link between target antigen density and effector mechanism efficacy, as quantified by flow cytometry in relapsed/refractory multiple myeloma cohorts treated with daratumumab monotherapy. Post-discontinuation, CD38 expression on hematopoietic cells, including NK cells and residual plasma cells, empirically recovers to baseline levels within approximately six months, indicating reversibility of the depletion effects. Prolonged daratumumab exposure, however, may contribute to NK cell exhaustion, as evidenced by phenotypic shifts and reduced functional recovery in long-term treated patients, potentially limiting sustained immune modulation.

Development and Regulatory Timeline

Discovery and Preclinical Studies

Daratumumab, a fully human IgG1κ monoclonal antibody targeting , was discovered by in the late 2000s using the company's HuMAb-Mouse platform for generating human antibodies. was selected as a target based on its overexpression on malignant plasma cells in , where it is present at high density on nearly all tumor cells, contrasted with low or absent expression on most normal hematopoietic cells, providing a favorable therapeutic window. This differential expression supported the rationale for antibody-mediated depletion of -positive myeloma cells while minimizing off-target effects on healthy tissues. Preclinical studies validated daratumumab's efficacy in vitro and in vivo, demonstrating multiple mechanisms of myeloma cell killing, including antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), antibody-dependent cellular phagocytosis (ADCP), and direct apoptosis induction. In mouse xenograft models using CD38-expressing human multiple myeloma cell lines, daratumumab monotherapy significantly inhibited tumor growth and induced regression, with ADCC identified as the predominant mechanism due to recruitment of natural killer cells. These findings, reported as early as 2009, confirmed the antibody's potency against primary myeloma cells and established proof-of-concept for CD38 targeting in hematologic malignancies. Genmab secured early intellectual property on daratumumab, including composition-of-matter patents extending into the 2020s, which facilitated progression to clinical development. Following these preclinical successes, Genmab filed an Investigational New Drug application with the U.S. FDA around 2010, enabling the initiation of first-in-human studies. In 2012, Genmab entered a global licensing agreement with Janssen Biotech, Inc., granting Janssen exclusive rights to further develop and commercialize the antibody while retaining milestones and royalties.

Pivotal Clinical Trials

The GEN501 trial, a phase 1/2 study initiated in 2008 and enrolling patients with relapsed or refractory multiple myeloma (RRMM), evaluated daratumumab monotherapy via dose escalation up to 24 mg/kg weekly, establishing the recommended dose of 16 mg/kg based on safety and preliminary efficacy. In the monotherapy cohort at 16 mg/kg (n=31 heavily pretreated patients), the overall response rate (ORR) was 29.2% (95% CI 20.8-38.9), with 2 complete responses and median duration of response of 7.6 months; however, the open-label design introduced potential assessment biases, and infusion-related reactions occurred in 56% of patients, mostly grade 1/2. These findings supported further development but highlighted limitations in durability for advanced disease. The phase 3 , randomized and open-label, enrolled 498 RRMM patients (1-3 prior lines) to plus bortezomib and dexamethasone (D-Vd) versus bortezomib-dexamethasone (Vd) from 2013 onward, with primary endpoint progression-free survival (PFS). Interim analysis showed median PFS not reached versus 7.2 months (HR 0.39, P<0.001), ORR 83% versus 63%, and later updates confirmed OS benefit (HR 0.71 after median 59.1 months follow-up), particularly in patients with one prior line. Open-label nature may have favored subjective endpoints like response rates, though blinded independent review mitigated some bias for PFS. Similarly, the phase 3 POLLUX trial (open-label, randomized) compared daratumumab plus lenalidomide and dexamethasone (D-Rd) versus Rd alone in 569 RRMM patients (≥1 prior line), demonstrating median PFS not reached versus 18.4 months (HR 0.37, P<0.001) and ORR 93% versus 82% in initial 2016 results. Four-year updates affirmed deepened responses and OS improvement (median 67.6 versus 52.5 months, HR 0.72). Limitations included higher-grade infections in the D-Rd arm and reliance on investigator-assessed endpoints prone to open-label bias. In newly diagnosed multiple myeloma (NDMM), the phase 3 MAIA trial (randomized, open-label) assessed D-Rd versus Rd in 737 transplant-ineligible patients, yielding median PFS 44.5 versus 17.5 months after 44.3 months follow-up (HR 0.47), ORR 93% versus 82%, and subsequent OS benefit (HR 0.66 at 5 years). The design's lack of blinding could inflate response durability estimates, though MRD negativity rates (28% versus 11% at 10^-5 sensitivity) supported biological activity.00466-6/fulltext) Recent phase 3 PERSEUS trial updates (randomized, open-label) in transplant-eligible NDMM (n=709) showed daratumumab-VRd induction/consolidation followed by D-R maintenance versus VRd yielding 48-month PFS of 84.5% versus 67.7% ( 0.42) and sustained MRD negativity in 64% of D-VRd patients associated with >95% PFS at 48 months. For high-risk smoldering myeloma, the phase 3 trial (randomized) compared daratumumab monotherapy (n=206) versus active monitoring, reducing progression or death risk by 51% ( 0.49; median follow-up 65.2 months) with ORR 63.4%, though long-term OS data remain immature and the population limits direct comparability to active MM trials.

Regulatory Approvals and Formulation Advances

Daratumumab received Designation from the () on May 1, 2013, for the treatment of in patients who had progressed after at least three prior therapies, including a and an immunomodulatory agent, or those to both classes. The granted accelerated approval on November 16, 2015, for intravenous daratumumab monotherapy in adults with relapsed or (RRMM) following four or more prior lines of therapy. The () followed with a conditional authorisation on May 20, 2016, for the same monotherapy indication in RRMM patients who had received at least three prior therapies. Label expansions occurred progressively for combination regimens in RRMM and newly diagnosed (NDMM). Between 2017 and 2023, FDA approvals included daratumumab with and dexamethasone (September 2017), and dexamethasone (January 2017, converted from accelerated to full), and dexamethasone (June 2019), and , , and for transplant-ineligible NDMM (June 2019). EMA approvals mirrored these for RRMM combinations by 2017-2019, with NDMM extensions including daratumumab--melphalan- in 2019 and further frontline combinations by 2023, though timings and exact regimens showed minor variances due to differing evidentiary thresholds. Post-approval commitments included evaluations for pediatric use, but the FDA waived such requirements in multiple instances, citing the rarity of in children and impracticability of studies. Formulation advances focused on administration efficiency. The FDA approved a split-dosing intravenous regimen on February 12, 2019, reducing infusion times after initial cycles. A subcutaneous , daratumumab and hyaluronidase-fihj (Darzalex Faspro), received FDA approval on May 1, 2020, enabling fixed-duration subcutaneous dosing in under five minutes for monotherapy and combinations across indications. The approved the subcutaneous version in June 2020. In 2025, the granted approval on July 23 for subcutaneous daratumumab monotherapy in high-risk smoldering , based on benefits. The FDA's Oncologic Drugs Advisory Committee voted favorably on May 20, 2025, supporting its benefit-risk profile for the same indication, with full approval pending. Global regulatory harmonization remains incomplete, with often adopting indications slightly ahead in some combination contexts while FDA emphasizes confirmatory data for conversions from accelerated status.

Economic and Access Issues

Pricing and Market Dynamics

In the United States, the wholesale acquisition cost for daratumumab (branded as Darzalex) stands at approximately $7,310 per 1,800 mg vial for the subcutaneous formulation, with intravenous doses typically requiring multiple vials for standard 16 mg/kg administrations, resulting in per-infusion costs of around $7,000 to $8,000 depending on patient weight and regimen. For regimens, such as daratumumab in combination with , , and dexamethasone, the drug acquisition cost reaches about $200,866 in the first year and $137,434 in the second year, contributing to annual expenses exceeding $300,000 when including full cycles. In the , pricing is negotiated lower through assessments, with cumulative vial costs ranging from €823 to €31,941 annually per patient, though exact figures vary by country and formulation; emerging markets see further reductions due to voluntary licensing and local manufacturing, often below EU levels but still substantial relative to GDP per capita. Patent exclusivity for , a , extends beyond initial composition-of-matter protections expiring around 2026-2029 in major markets, with additional formulation and method-of-use pushing full entry into the 2030s, including potential U.S. protections until 2035. This prolonged exclusivity supports Janssen's () market dominance, where Darzalex generated $11.6 billion in global sales in recent years, capturing a leading share—estimated over 70%—of the through expanded indications and combination therapies. Discounts negotiated through pharmacy benefit managers (PBMs) and the 340B program substantially lower net prices from list levels, with reporting $47.8 billion in total rebates, fees, and discounts across its portfolio in 2024, including growing 340B payments of $7.4 billion. However, transparency reports indicate that 340B entities often apply markups of up to 4.9 times acquisition costs on drugs like daratumumab, sustaining high net expenditures despite these mechanisms, as gross-to-net reductions reached $334 billion industry-wide for brand-name drugs in 2023. Market dynamics are further driven by Janssen's subcutaneous formulation advancements, which reduce administration times and costs compared to intravenous versions, bolstering uptake without eroding .

Cost-Effectiveness Evaluations

Cost-effectiveness analyses of daratumumab in relapsed/refractory (RRMM) have generally reported incremental cost-effectiveness ratios (ICERs) exceeding $200,000 per quality-adjusted life-year (QALY) gained, despite modest QALY increments of 1-2 years. For instance, in comparisons of daratumumab plus and dexamethasone (DRd) versus alternative regimens like VRd or Rd, models yielded ICERs of approximately $1.4 million per progression-free QALY, driven by high drug acquisition costs that outweighed extensions in (PFS). Similarly, evaluations of daratumumab plus , , and (D-VMP) in transplant-ineligible patients showed additional QALYs of about 2 but ICERs around $226,000-262,000, with analyses highlighting vulnerability to discount rates and assumptions about development. In newly diagnosed (NDMM), particularly high-risk or transplant-eligible subsets, modeling based on the trial suggests more favorable outcomes. Extrapolations from data, which demonstrated superior PFS and negativity with daratumumab plus , , and dexamethasone (D-VRd) versus VRd, indicate potential QALY gains supporting cost-effectiveness under certain thresholds, especially when factoring in reduced need for subsequent therapies and long-term PFS projections. One analysis affirmed first-line daratumumab as cost-effective in transplant-eligible NDMM, contrasting with broader first-line critiques where ICERs exceeded payer thresholds like $150,000/QALY due to upfront costs. High daratumumab acquisition costs—often $6,500 per —causally offset PFS benefits in base-case models, with ICERs proving sensitive to weights, time horizons, and rates that diminish long-term gains. Internationally, variances arise from negotiated pricing in single-payer systems; for example, impact models in settings with volume-based discounts have projected daratumumab as cost-effective at willingness-to-pay thresholds below U.S. levels, though probabilistic analyses show in 40-50% of scenarios.

Global Access and Biosimilar Prospects

Daratumumab has secured regulatory approvals in numerous countries worldwide, including the , nations, , , and others, enabling its integration into treatment protocols since its initial FDA approval in 2015. By October 2024, over 500,000 patients globally had received daratumumab-based therapies, reflecting broad dissemination in high-income markets. However, penetration remains low in low- and middle-income countries (LMICs), where economic constraints limit adoption despite the persistence of older regimens like and ; a 2023 international survey found daratumumab available but underutilized in such settings due to affordability barriers. The has evaluated daratumumab for its Model List of but declined inclusion, with expert committees highlighting insufficient evidence of cost-effectiveness for equitable access in resource-limited environments as of the 2023 list. Reimbursement for daratumumab varies significantly by region, influencing real-world uptake. In the United States, Part B covers the drug for FDA-approved indications following initial approvals, subject to standard deductibles and copayments. Globally, assessments (HTAs) in cost-constrained systems have led to rejections or restrictions; for instance, Australia's denied public funding for frontline daratumumab combinations in 2023, citing inadequate value relative to incremental benefits. Such decisions underscore disparities, with high-income countries achieving higher rates while LMICs rely on out-of-pocket payments or forego the altogether. Prospects for biosimilars hinge on patent expirations and regulatory hurdles for complex monoclonal antibodies like daratumumab. Key patents are expected to lapse starting in 2029 in the , 2030 in , and 2031 in , potentially paving the way for biosimilar competition post-2030 in these markets. As of October 2025, no s are approved in the or , though an active pipeline includes candidates such as HLX15 (developed by Shanghai Henlius Biotech in partnership with ), with clinical development ongoing for indications. The first daratumumab , Daratumia by BIOCAD, gained approval in in August 2025, marking an early entry in select jurisdictions and hinting at gradual expansion to enhance affordability where originator pricing dominates. These advancements could mitigate access gaps, particularly if WHO prequalification facilitates quality-assured versions for LMICs.

Criticisms and Limitations

Debates on Clinical Value

Daratumumab has demonstrated improved (PFS) and overall survival (OS) in randomized controlled trials for , particularly when added to backbone regimens, with proponents citing deep responses and high rates of (MRD) negativity as evidence of substantial clinical value. In the trial for transplant-ineligible newly diagnosed patients, daratumumab plus and dexamethasone (D-Rd) yielded a OS of 67.6 months compared to 51.8 months with lenalidomide and dexamethasone alone, representing an approximate 16-month absolute gain. Pooled analyses from trials in relapsed/refractory settings showed OS of 65.0 months with daratumumab versus 38.2 months with control arms. These outcomes are supported by elevated MRD negativity rates, such as 14.9% sustained for at least 6 months with D-Rd versus 4.3% in controls, which correlate with prolonged PFS and OS in multiple studies. Advocates argue these metrics justify daratumumab's frontline positioning, as MRD negativity serves as a prognostic indicator of durable remission. However, skeptics question the robustness of these benefits, noting that surrogate endpoints like MRD negativity and PFS do not always reliably predict OS gains, with meta-analyses showing only moderate associations between MRD status and OS due to effective subsequent therapies mitigating differences. Regulatory bodies have not fully endorsed MRD as a validated for OS, citing methodological variability across trials that limits its beyond PFS. In , PFS durations are often shorter than in trials, particularly with prior exposures; for instance, median PFS was 19.8 months for patients with 0-1 prior lines versus 6.2 months for those with 2 or more, reflecting challenges in heavily pretreated populations where trial optimism from press releases may not translate. Absolute OS extensions in some relapsed settings, such as 12.4 months median PFS in the APOLLO trial, are viewed as modest increments over historical standards rather than transformative shifts. Further debate surrounds daratumumab's incremental value atop modern regimens, where additions yield PFS improvements but OS data remain immature or show hazard ratios suggesting relative rather than absolute dominance; for example, quadruplet regimens with daratumumab enhance response depth over like VRd, yet critics contend the gains—such as reduced progression risk without proportional OS leaps—may not outweigh positioning it early, preserving it for later lines amid evolving therapies. While superior to historical controls, real-world variability and surrogate-to-hard-outcome discrepancies prompt caution against over-positioning based solely on endpoints.

Resistance Mechanisms and Durability

Resistance to daratumumab in arises primarily through antigen escape, where malignant plasma cells downregulate or lose expression on their surface, enabling evasion of (ADCC), (CDC), and direct induced by the drug. This loss often results from selective pressure favoring pre-existing -low subclones or post-treatment downregulation via (transfer of -daratumumab complexes to effector cells like monocytes and granulocytes) and upregulation of complement inhibitors such as CD55 and CD59. Clonal evolution under daratumumab therapy thus promotes outgrowth of resistant variants, mirroring immune evasion patterns observed with other monoclonal antibodies targeting tumor antigens. In relapsed or following prior daratumumab exposure, overall response rates to retreatment or subsequent anti-CD38 regimens typically range from 30% to 54%, reflecting diminished efficacy due to these escape mechanisms, with median often limited to 6-8 months in refractory cohorts. Combination therapies, such as daratumumab with lenalidomide-dexamethasone or , can partially mitigate resistance by engaging complementary pathways (e.g., or inhibition), yet empirical data indicate median durations of response or plateau at approximately 12-24 months in relapsed settings, even with extended follow-up through 2025. No clinically validated biomarkers reliably predict primary resistance to daratumumab, though exploratory studies highlight potential roles for baseline expression levels, NK cell phenotypes, and genomic alterations in the bone marrow microenvironment; these factors influence susceptibility but lack prospective utility for patient selection. The absence of such markers underscores the challenge of indefinite daratumumab use, as cumulative immune adaptations and tumor heterogeneity inevitably erode long-term control, necessitating sequential therapies.

Overuse and Resource Allocation Concerns

Daratumumab's expansion into earlier lines of therapy for has prompted concerns over "line creep," where use precedes robust overall survival data from pivotal trials initially focused on relapsed or settings. A 2024 payer analysis determined that incorporating daratumumab in first-line treatment yields an exceeding $150,000 per quality-adjusted life-year , highlighting marginal clinical gains relative to escalating expenditures. This shift diverts finite healthcare resources from lower-cost generics or supportive options, particularly in settings where proportional survival extensions remain unproven against established backbones like lenalidomide-dexamethasone. Real-world utilization patterns reveal frequent deviations from approved dosing schedules, with daratumumab administered more often than FDA recommendations, thereby inflating costs without of enhanced . Such overuse contributes to substantial budget strains; for instance, integrating daratumumab into relapsed-refractory regimens is projected to raise healthcare expenditures by $6.17 million over five years in modeled populations adopting it at scale. In resource-constrained systems, this imposes opportunity costs, prioritizing high-cost biologics over palliative or generic alternatives where causal chains link expenditures to limited incremental benefits in rather than transformative outcomes. Critics argue that pharmaceutical-sponsored expansions, often via combination approvals, outpace conservative application in lower-risk or ineligible patients, as real-world data underscore higher-than-necessary utilization without commensurate durability gains. Registries and pharmacoeconomic models emphasize reserving daratumumab for later lines where is strongest, avoiding non-evidence-based creep that burdens systems amid stagnant overall curves in early adoption scenarios. This perspective aligns with causal assessments prioritizing empirical thresholds for , cautioning against routine frontline deployment absent proportional value in diverse patient cohorts.

References

  1. [1]
    Daratumumab: a first-in-class CD38 monoclonal antibody for ... - NIH
    Jun 30, 2016 · Preclinical studies have shown that daratumumab induces MM cell death through several mechanisms, including complement-dependent cytotoxicity ( ...Missing: peer | Show results with:peer
  2. [2]
    CD38 Antibodies in Multiple Myeloma: Mechanisms of Action and ...
    Daratumumab reduces CD38 on the cell surface by several mechanisms. First, in responding patients daratumumab may select for MM cells with lower CD38 expression ...
  3. [3]
    Daratumumab (DARZALEX) - FDA
    Nov 22, 2016 · The US Food and Drug Administration approved daratumumab (DARZALEX, Janssen Biotech, Inc.) in combination with lenalidomide and dexamethasone, or bortezomib ...
  4. [4]
    FDA approves daratumumab for multiple myeloma ineligible for ...
    Jun 28, 2019 · The Food and Drug Administration approved daratumumab (DARZALEX, Janssen Biotech, Inc.) in combination with lenalidomide and dexamethasone for patients with ...
  5. [5]
    [PDF] Darzalex - accessdata.fda.gov
    ... daratumumab is a protein. Do not use if visibly opaque ... consider using a FDA-approved daratumumab-specific IFE assay to distinguish daratumumab.
  6. [6]
    FDA approves daratumumab and hyaluronidase-fihj for multiple ...
    May 1, 2020 · The recommended dose of daratumumab and hyaluronidase-fihj is 1,800 mg daratumumab and 30,000 units hyaluronidase administered subcutaneously ...
  7. [7]
    Evaluating Daratumumab in the Treatment of Multiple Myeloma - NIH
    Aug 26, 2020 · Another distinct mechanism is its immunomodulatory action. Specifically, daratumumab binds to the CD38 positioned on the surface of immune ...Missing: peer | Show results with:peer
  8. [8]
    Darzalex | European Medicines Agency (EMA)
    At the time of approval, Darzalex was the first medicine authorised for the treatment of smouldering multiple myeloma. Darzalex's side effects are considered ...Overview · Product details · Authorisation details · Assessment history
  9. [9]
    Darzalex (daratumumab) FDA Approval History - Drugs.com
    Sep 30, 2019 · FDA approval history for Darzalex (daratumumab) used to treat Multiple Myeloma. Supplied by Janssen Biotech, Inc.
  10. [10]
    FDA Approves Daratumumab in Combination for Frontline Multiple ...
    On September 26, 2019, the U.S. Food and Drug Administration (FDA) approved daratumumab (Darzalex, Janssen) for adult patients with multiple myeloma in ...
  11. [11]
    FDA approves daratumumab and hyaluronidase-fihj with bortezomib ...
    Jul 30, 2024 · The recommended Darzalex Faspro dose is 1,800 mg/30,000 units (1,800 mg daratumumab and 30,000 units hyaluronidase). See the prescribing ...
  12. [12]
    DARZALEX® (daratumumab)-SC based quadruplet regimen ...
    Oct 23, 2024 · DARZALEX® (daratumumab)-SC based quadruplet regimen approved by the European Commission for patients with newly diagnosed multiple myeloma who ...Missing: Rd | Show results with:Rd
  13. [13]
    European Commission approves DARZALEX® (daratumumab) as ...
    Jul 23, 2025 · Daratumumab is the only CD38-directed antibody approved to be given subcutaneously to treat patients with multiple myeloma.19 Daratumumab SC is ...
  14. [14]
    FDA ODAC Supports Use of Darzalex Faspro for HR-SMM
    Jun 5, 2025 · According to applicant Johnson & Johnson, results of the AQUILA study demonstrated that early with daratumumab helped patients with high-risk ...
  15. [15]
    Four-Year Follow-up of the Phase 3 Pollux Study of Daratumumab ...
    Nov 13, 2019 · D-Rd was associated with a significantly higher ORR versus Rd (93% vs 76%), including higher rates of ≥very good partial response (81% vs 49%) ...
  16. [16]
    subgroup analysis of the phase 3 CASTOR and POLLUX studies - NIH
    In POLLUX, daratumumab in combination with Rd (D-Rd) reduced the risk of disease progression or death by 63% compared with Rd after a median follow-up of 13.5 ...
  17. [17]
    Daratumumab, Bortezomib, and Dexamethasone Versus ...
    The 42-month PFS rates were 22% for D-Vd and 1% for Vd. For patients with 1 to 3 prior lines of therapy, median PFS was 18.0 months with D-Vd versus 7.3 months ...
  18. [18]
    New long-term progression free survival data projections reinforce ...
    Apr 23, 2025 · Median progression free survival projected to be 17 years for transplant eligible patients receiving daratumumab-based regimen.
  19. [19]
    Daratumumab, Bortezomib, Lenalidomide, and Dexamethasone for ...
    Daratumumab has been approved for use in combination with various regimens for the treatment of patients with newly diagnosed multiple myeloma, including a ...
  20. [20]
    Daratumumab-based quadruplet therapy for transplant-eligible ...
    Apr 22, 2024 · Here, we present a post hoc analysis of patients with high-risk cytogenetic abnormalities (HRCAs; del[17p], t[4;14], t[14;16], t[14;20], or gain ...
  21. [21]
    Daratumumab or Active Monitoring for High-Risk Smoldering ...
    Despite some evidence of benefit, these trials did not result in approved treatments for high-risk smoldering multiple myeloma. In AQUILA, we found a ...
  22. [22]
    Daratumumab Shows Clinical Activity, No New Safety Concerns ...
    Jun 16, 2025 · Daratumumab Shows Clinical Activity, No New Safety Concerns With Extended Use in Intermediate/High-Risk Smoldering MM Available. June 16, 2025.
  23. [23]
    Daratumumab-Based Therapeutic Approaches and Clinical ...
    Oct 11, 2024 · Newly diagnosed and relapsed/refractory patients had an ORR of 92% and 76.3%, and a ≥ VGPR (very good partial response) rate of 80% and 55.3%, ...
  24. [24]
    Real-World Evidence of Daratumumab Monotherapy in Relapsed ...
    Overall response rate (ORR) was 30% with positive impact on progression-free survival (PFS).
  25. [25]
    Real-world analysis of treatment patterns, effectiveness, and safety ...
    May 7, 2025 · Daratumumab is a human IgGκ monoclonal antibody targeting CD38 with direct on-tumor and immunomodulatory mechanisms of action.
  26. [26]
    Three-year Follow-up of CASTOR
    Oct 9, 2019 · With extended follow-up, significant improvements in PFS, ORR, and MRD negativity were observed for patients with RRMM treated with D-Vd versus ...
  27. [27]
    A Systematic Literature Review and Meta-Analysis - PubMed
    This study aimed to systematically identify evidence on the clinical effectiveness of DRd and VRd as first-line treatments for patients with TIE NDMM and to ...Missing: pomalidomide | Show results with:pomalidomide
  28. [28]
    Real-world comparison of daratumumab-based regimens in ...
    Mar 8, 2024 · Daratumumab (dara)-based triplet therapies are commonly used in the second-line (2L) and third-line (3L) settings in relapsed/refractory multiple myeloma (RRMM ...
  29. [29]
    Comparative Efficacy of Daratumumab Monotherapy and ... - NIH
    The MAIC results suggest that daratumumab improves OS compared with pomalidomide + LoDex in patients with heavily pretreated multiple myeloma.
  30. [30]
    Progression-Free Survival of Daratumumab Versus Bortezomib ...
    DRd is associated with a significantly lower risk of disease progression or death compared to VRd as 1L treatment for TIE NDMM patients.
  31. [31]
    Daratumumab-based immunotherapy vs. lenalidomide, bortezomib ...
    This meta-analysis suggests that daratumumab-containing immunotherapy is superior to RVD in the depth of treatment efficacy, progression-free survival, and ...
  32. [32]
    What Are the Most Effective Therapies for Daratumumab-Refractory ...
    Mar 18, 2025 · A systematic review found that CAR T-cell therapy had the best overall survival and progression-free survival in patients with multiple ...
  33. [33]
    Treatment of Multiple Myeloma in Patients Refractory to ...
    Mar 7, 2025 · This review aims to identify and compare treatments with published clinical trial evidence among patients with daratumumab‐refractory multiple myeloma.
  34. [34]
    [PDF] Comparative Efficacy of CARVYKTI in CARTITUDE-4 versus ...
    Sep 28, 2024 · Cilta-cel showed superior efficacy compared to alternative treatments from the daratumumab clinical trials, across all outcomes (ORR, ≥VGPR,. ≥ ...
  35. [35]
    A network meta-analysis of randomized clinical trials in lenalidomide ...
    Jul 15, 2025 · Our findings indicate that belantamab mafodotin, bortezomib, and dexamethasone (BVd) combination is the most effective regimen for both ...
  36. [36]
    Final analysis of the phase III non-inferiority COLUMBA study of ...
    Mar 31, 2022 · In summary, DARA SC and DARA IV continue to demonstrate similar efficacy and safety, with a low rate of infusion-related reactions (12.7% vs.
  37. [37]
    Practical Considerations for the Daratumumab Management in ... - NIH
    The overall risk of infection with daratumumab treatment is around 38%, being upper respiratory tract infections the most common.
  38. [38]
    DARZALEX - Management of Infusion-Related Reactions
    Aug 1, 2025 · DARZALEX is contraindicated in patients with a history of severe hypersensitivity (e.g., anaphylactic reactions) to daratumumab or any of the ...
  39. [39]
    Practical Considerations for Antibodies in Myeloma
    May 23, 2018 · Use of prophylactic antibiotics is recommended for patients who receive either daratumumab or elotuzumab. This prophylaxis should include an ...
  40. [40]
    Management of infectious risk of daratumumab therapy in multiple ...
    However, the addition of daratumumab to established regimens for the treatment of MM has an increased risk of adverse events. An integrated safety analysis ...
  41. [41]
    darzalex faspro - Hepatitis B Virus Reactivation - J&J Medical Connect
    Mar 26, 2025 · SUMMARY. Hepatitis B virus (HBV) reactivation, in some cases fatal, has been reported in patients treated with DARZALEX for intravenous (IV) ...
  42. [42]
    Reactivation of resolved hepatitis B after daratumumab for multiple ...
    Apr 13, 2021 · Among 93 patients with daratumumab treatment, reactivation occurred in 6 patients (6.5%) with one hepatic failure. This is the first report ...
  43. [43]
    [PDF] 1088 or www.fda.gov/medwatch. Reference ID: 5516922
    DARZALEX can cause severe and/or serious infusion-related reactions including anaphylactic reactions.
  44. [44]
    Prevention and Management of Daratumumab Mediated Infusion ...
    Nov 13, 2019 · Daratumumab is associated with infusion related reactions (IRRs), which present with symptoms of rhinitis, cough, dyspnea, bronchospasm, chills and nausea.
  45. [45]
    Daratumumab/lenalidomide/dexamethasone in transplant-ineligible ...
    Feb 27, 2025 · Rate of daratumumab discontinuation due to TEAEs was 14.6%. ... Longer term follow-up of the randomized phase III trial SWOG S0777 ...
  46. [46]
    Updates in Multiple Myeloma from ASH 2024 - MMRF
    Safety: Grade 3/4 AEs were more common with daratumumab (40.4% vs 30.1%), but the treatment was generally well tolerated, with low discontinuation rates (5.7%).
  47. [47]
    6 Year Update of an Open-label, Randomised, Phase 3 Trial - PMC
    Aug 25, 2025 · Daratumumab maintenance significantly improved progression-free survival and increased minimal residual disease (MRD)-negativity rates versus ...
  48. [48]
    New analyses reinforce long-term benefit of DARZALEX ...
    Jun 3, 2025 · Sustained MRD negativity (10-5) rates at 24 months or longer were more than doubled in transplant eligible patients treated with ...Missing: cytopenias | Show results with:cytopenias
  49. [49]
    Daratumumab with lenalidomide as maintenance after transplant in ...
    Jan 16, 2025 · Incidences of grade 3/4 cytopenias (54.2% vs 46.9%) and infections (18.8% vs 13.3%) were slightly higher with D-R than R. In conclusion, D-R ...
  50. [50]
    Cumulative Incidence and Relative Risk of Infection in Patients With ...
    A number of studies have suggested a connection between daratumumab treatment and hypogammaglobulinemia, which could result in increased risk for infection ...
  51. [51]
    Efficacy and Safety of Daratumumab, Pomalidomide and ...
    Aug 27, 2025 · Both treatment strategies demonstrated comparable survival outcomes, with no statistically significant differences in progression-free survival ...Baseline Patient... · Prognostic Factors For... · Overall Survival And...
  52. [52]
    Real-World Effectiveness and Safety of Intravenous Daratumumab ...
    The aim of this observational study was to evaluate the effectiveness and safety of daratumumab in real-world clinical practice.
  53. [53]
    Clinical Pharmacokinetics and Pharmacodynamics of Daratumumab
    At a median follow-up of approximately three years, the most common (>20%) treatment-emergent adverse events were fatigue (42%), nausea (30%), anemia (28%), ...
  54. [54]
    [PDF] DARZALEX PI
    Feb 27, 2025 · No drug-drug interaction studies have been performed. Clinical pharmacokinetic assessments of daratumumab in combination with lenalidomide,.
  55. [55]
    [PDF] 3629-Multiple myeloma daratumumab maintenance | eviQ
    No drug-drug interaction studies have been performed. Clinical pharmacokinetic assessments of daratumumab in combination with lenalidomide, pomalidomide ...
  56. [56]
    [PDF] 761145Orig1s000 - accessdata.fda.gov
    Feb 25, 2021 · The purpose of adding hyaluronidase, an endoglycosidase, to daratumumab DP is to increase the dispersion and absorption of daratumumab when ...
  57. [57]
    Subcutaneous daratumumab and hyaluronidase-fihj in newly ...
    In its subcutaneous formulation, daratumumab is combined with recombinant human hyaluronidase PH20 (rHuPH20; ENHANZE drug delivery technology; Halozyme, Inc.
  58. [58]
    Target-Mediated Drug Disposition of Daratumumab Following ...
    Age, race, renal impairment, and mild hepatic impairment had neither statistically significant nor clinically relevant effects on exposure to daratumumab.
  59. [59]
    [PDF] Darzalex, INN - daratumumab - European Medicines Agency
    Based on population pharmacokinetic (PK) analyses no dose adjustment is necessary for patients with renal impairment (see section 5.2). Hepatic impairment. No ...
  60. [60]
    Interference of daratumumab with pretransfusion testing, mimicking ...
    The presence of daratumumab in the sera can interfere with pretransfusion testing due to the weakly expression of CD38 on red cells.
  61. [61]
    Interaction with Serological Testing for Blood Banks
    Daratumumab binds to CD38 found at low levels on red blood cells (RBCs) and may result in a positive indirect Coombs test (indirect antiglobulin test; IAT).
  62. [62]
    [PDF] Understanding Daratumumab Interference With Blood Compatibility ...
    Daratumumab does not interfere with identification of ABO/RhD antigens. • If an emergency transfusion is required, noncrossmatched, ABO/RhD-compatible RBCs ...
  63. [63]
    Optimized strategy to mitigate daratumumab interference in blood ...
    Jul 2, 2025 · This occurs due to DARA binding to CD38 expressed on the surface of red blood cells (RBCs).
  64. [64]
    Validation of Daraex to Resolve Daratumumab-Induced ...
    Nov 13, 2019 · This interference hinders the detection of red blood cell alloantibodies. Published literature has described a method to eliminate CD38 in red ...
  65. [65]
    [PDF] Mitigating the Anti-CD38 Interference with Serologic Testing - AABB
    Jan 15, 2016 · International validation of a dithiothreitol. (DTT)-based method to resolve the daratumumab interference with blood compatibility testing ...
  66. [66]
    ANTI-CD38 NANOBODY JK36 ALLOWS RELIABLE MRD ... - NIH
    Jun 23, 2022 · In flow cytometry CD38 is frequently used as a gating marker of plasma cells (PCs), but daratumumab can hampering myeloma cell detection. A ...
  67. [67]
    Flow cytometric myeloma measurable residual disease testing in the ...
    Jul 20, 2021 · This review will focus on the intersection of MM MRD testing and the advent of MM targeted therapies. 2 ANTI-CD38 THERAPY IN MYELOMA.
  68. [68]
    Effects of Daratumumab (Anti-CD38) Monoclonal Antibody Therapy ...
    This study highlights a significant interference caused by Daratumumab in the flow cytometry evaluation of samples for minimal residual disease (MRD) testing in ...
  69. [69]
    Daratumumab: Dawn of a New Paradigm in Multiple Myeloma? - PMC
    MECHANISM OF ACTION. Daratumumab is a human immunoglobulin (IgG1) monoclonal antibody directed against CD38, which is highly expressed on myeloma cells. It ...
  70. [70]
    Daratumumab and its potential in the treatment of multiple myeloma
    Daratumumab is a novel, high-affinity, therapeutic human monoclonal antibody against unique CD38 epitope with broad-spectrum killing activity.
  71. [71]
    Mechanisms of Resistance to Anti-CD38 Daratumumab in Multiple ...
    Jan 9, 2020 · Dara exerts anti-MM activity via antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), ...
  72. [72]
    CD38 expression and complement inhibitors affect response and ...
    We have previously shown that expression of CD38 on MM cells correlates with daratumumab-mediated ADCC and CDC in vitro. ... apoptosis of myeloma tumor ...Cd38 Expression Levels On... · Reduced Cd38 Expression On... · Increased Cd55 And Cd59...
  73. [73]
    CD38 Antibodies in Multiple Myeloma: Mechanisms of Action and ...
    Sep 20, 2018 · Reduced CD38 expression on non-depleted MM cells is associated with protection against ADCC and CDC (63, 68). Reduced daratumumab-mediated ADCC ...Cd38 Target Antigen · Cd38 And Primary Resistance · Cd38 And Acquired Resistance
  74. [74]
    Pharmacokinetics of Daratumumab Following Intravenous Infusion ...
    Nov 29, 2016 · Following intravenous infusion, daratumumab exhibits nonlinear pharmacokinetic characteristics, consistent with target-mediated drug disposition ...
  75. [75]
    [PDF] Reference ID: 4947469 - accessdata.fda.gov
    Daratumumab-mediated positive indirect antiglobulin test may persist for up to 6 months after the last daratumumab infusion.
  76. [76]
    Darzalex Faspro: Package Insert / Prescribing Information - Drugs.com
    Aug 12, 2024 · The recommended dose of DARZALEX FASPRO is 1,800 mg/30,000 units (1,800 mg daratumumab and 30,000 units hyaluronidase) administered ...<|separator|>
  77. [77]
    DARZALEX - Pharmacokinetics - J&J Medical Connect
    Nov 21, 2024 · Renal excretion and hepatic enzyme-mediated metabolism of intact daratumumab are therefore unlikely to represent major elimination routes.
  78. [78]
    Darzalex Faspro FDA Approval History - Drugs.com
    Jul 31, 2024 · Darzalex Faspro FDA Approval History ; FDA Approved: Yes (First approved May 1, 2020) ; Brand name: Darzalex Faspro ; Generic name: daratumumab and ...
  79. [79]
    Exposure‐Response and Population Pharmacokinetic Analyses of a ...
    Nov 3, 2020 · The estimated bioavailability for subcutaneous DARA was approximately 70%. The estimated linear clearance was 0.005 L/h, and the volume of ...
  80. [80]
    Clinical Administration Characteristics of Subcutaneous and ...
    Feb 9, 2023 · Median total chair times were 2.7-2.8 hours shorter for DARA SC versus DARA IV. Incidences of ARR-related events with DARA SC were low across ...
  81. [81]
    CD38 antibodies in multiple myeloma: back to the future
    Jan 4, 2018 · Similarly, daratumumab-treated patients who achieved at least PR, had higher baseline CD38 expression on their tumor cells, compared with ...Missing: density | Show results with:density
  82. [82]
    Effects of daratumumab on natural killer cells and impact on clinical ...
    Oct 24, 2017 · Daratumumab, like other CD38 antibodies, reduced NK-cell counts in peripheral blood mononuclear cells (PBMCs) of healthy donors in vitro.
  83. [83]
    CD38 Expression Levels Linked to Daratumumab Response in ...
    Jun 22, 2016 · CD38 expression was associated with response to daratumumab monotherapy in patients with multiple myeloma.
  84. [84]
    NK Cell Phenotype Is Associated With Response and Resistance
    First, the efficacy of daratumumab has been shown to be partially dependent on baseline CD38 expression on MM cells. 13 However, the role of CD38 in acquired ...
  85. [85]
    Daratumumab: First Global Approval - PubMed
    Daratumumab (Darzalex™) is a first-in-class, humanized IgG1κ monoclonal antibody that targets the CD38 epitope and was developed by Janssen Biotech and Genmab.
  86. [86]
    Upregulation of CD38 expression on multiple myeloma cells by ...
    Apr 29, 2020 · CD38 is highly and ubiquitously expressed on MM cells and at low levels on normal lymphoid and myeloid cells [8]. CD38 is a transmembrane ...
  87. [87]
    Preclinical Evidence for the Therapeutic Potential of CD38-Targeted ...
    The human CD38 monoclonal antibody daratumumab is currently being evaluated in phase I/II clinical trials as a novel therapy for multiple myeloma. Here, we ...
  88. [88]
    Daratumumab: a first-in-class CD38 monoclonal antibody for the ...
    Jun 30, 2016 · Given that daratumumab induces ADCC by NK cells and lenalidomide promotes NK cell activity, van der Veer et al. investigated daratumumab ...
  89. [89]
    Daratumumab, a Novel Potent Human Anti-CD38 Monoclonal ...
    Nov 20, 2009 · Daratumumab, a novel potent human anti-CD38 monoclonal antibody, induces significant killing of human multiple myeloma cells: therapeutic implication.
  90. [90]
    securities and exchange commission - SEC Filing - Genmab A/S
    Our issued U.S., European and Japanese patents covering the composition of matter for daratumumab do not begin to expire until March 2026. In September 2020, ...
  91. [91]
    Daratumumab - New Drug Approvals
    Apr 26, 2016 · Genmab announced a global license and development agreement for daratumumab with Janssen Biotech, Inc. ... IND Filed · INDIA 2020 · INDIA 2022 ...
  92. [92]
    Janssen Biotech Announces Global License and Development ...
    Aug 30, 2012 · Genmab will grant Janssen an exclusive worldwide license to develop and commercialize daratumumab as well as a back-up CD38 human antibody.Missing: IND | Show results with:IND
  93. [93]
    Daratumumab (HuMax®-CD38) Safety Study in Multiple Myeloma
    This study is conducted in two parts. In part I, participants are enrolled into cohorts at increasing dose levels. Participant safety and efficacy during ...Missing: pivotal | Show results with:pivotal
  94. [94]
    EMA Review of Daratumumab for the Treatment of Adult Patients ...
    Daratumumab monotherapy achieved an overall response rate of 29.2% (95% confidence interval [CI] 20.8 to 38.9) in patients with multiple myeloma who had ...
  95. [95]
    final results from the phase 2 GEN501 and SIRIUS trials - PubMed
    Daratumumab showed encouraging efficacy as monotherapy in patients with heavily pretreated multiple myeloma in the GEN501 and SIRIUS studies.Missing: pivotal | Show results with:pivotal
  96. [96]
    Study Details | NCT02136134 - Clinical Trials
    Overall Survival With Daratumumab, Bortezomib, and Dexamethasone in Previously Treated Multiple Myeloma (CASTOR): A Randomized, Open-Label, Phase III Trial.
  97. [97]
    Daratumumab, Bortezomib, and Dexamethasone for Multiple Myeloma
    Aug 25, 2016 · We report the results of a prespecified interim analysis of a randomized phase 3 trial of daratumumab in combination with bortezomib and ...
  98. [98]
    A Randomized, Open-Label, Phase III Trial - PubMed
    Mar 10, 2023 · D-Vd significantly prolonged OS in patients with RRMM, with the greatest OS benefit observed in patients with one prior line of therapy.
  99. [99]
    results from the phase III CASTOR trial - PubMed
    Feb 8, 2021 · In the phase III CASTOR trial, daratumumab, bortezomib and dexamethasone (D-Vd) significantly extended progression-free survival compared with bortezomib and ...
  100. [100]
    Daratumumab, Lenalidomide, and Dexamethasone for Multiple ...
    Oct 6, 2016 · The most common adverse events leading to death were acute kidney injury (in 0.4% of the patients in the daratumumab group and in 1.1% in the ...<|separator|>
  101. [101]
    Overall Survival With Daratumumab, Lenalidomide, and ...
    Jan 4, 2023 · POLLUX was a multicenter, randomized, open-label, phase III study during which eligible patients with ≥ 1 line of prior therapy were randomly ...
  102. [102]
    Daratumumab plus Lenalidomide and Dexamethasone for ...
    May 29, 2019 · After a median follow-up of 44.3 months, the median progression-free survival was 44.5 months in the daratumumab group, as compared with 17.5 ...Missing: pivotal | Show results with:pivotal
  103. [103]
    overall survival results from a randomised, open-label, phase 3 trial
    Daratumumab plus lenalidomide and dexamethasone increased overall survival and progression-free survival in patients ineligible for stem-cell transplantation ...
  104. [104]
    Analysis of sustained minimal residual disease negativity in the ...
    May 28, 2025 · In TE NDMM, nearly two-thirds of pts treated with DVRd induction and DR maint achieved ≥12-mo sustained MRD neg, associated with >95% 48-mo PFS rate.
  105. [105]
    DARZALEX FASPRO® (daratumumab and hyaluronidase-fihj)
    PERSEUS is an ongoing, randomized, open-label, Phase 3 study comparing the efficacy and safety of D-VRd during induction and consolidation versus VRd during ...
  106. [106]
    Daratumumab or Active Monitoring for High-Risk Smoldering ...
    With a median follow-up of 65.2 months, the risk of disease progression or death was 51% lower with daratumumab than with active monitoring (hazard ratio, 0.49; ...
  107. [107]
  108. [108]
    [PDF] bla 761145/s-007 supplement approval - accessdata.fda.gov
    Jul 9, 2021 · We are waiving the pediatric study requirement for this application because necessary studies are impossible or highly impracticable.
  109. [109]
    U.S. FDA Approves DARZALEX® (daratumumab) Split-Dosing ...
    Feb 12, 2019 · The most frequent serious adverse reactions were pneumonia (6%), general physical health deterioration (3%), and pyrexia (3%). Treatment- ...<|separator|>
  110. [110]
    Genmab Announces European Marketing Authorization for the ...
    Jun 4, 2020 · The approval follows a Positive Opinion by the CHMP of the European Medicines Agency (EMA) in April 2020. The SC formulation is administered as ...
  111. [111]
    U.S. FDA Oncologic Drugs Advisory Committee votes in favor of the ...
    May 20, 2025 · Daratumumab may increase thrombocytopenia induced by background therapy. Monitor complete blood cell counts periodically during treatment ...
  112. [112]
    Table 4, Cost and Cost-Effectiveness - Daratumumab (Darzalex SC)
    Table 4Cost and Cost-Effectiveness ; Submitted price, Daratumumab SC: $7,310 per 1,800 mg vial ; Treatment cost, Recommended dosage: Cycle 1 and cycle 2: 1,800 mg ...
  113. [113]
  114. [114]
    Cost of Anti-CD38 Monoclonal Antibodies in Combination With ...
    Jul 21, 2025 · The DAC was $200 866 in year 1 and $137 434 in year 2 for daratumumab and $212 421 in year 1 and $144 143 in year 2 for isatuximab. The DAC of ...
  115. [115]
    A case study for pembrolizumab and daratumumab - PMC - NIH
    Feb 1, 2024 · The yearly cumulative cost-based prices (CBPs) ranged from €52 to €885 for pembrolizumab per vial and €823 to €31,941 for daratumumab per vial.
  116. [116]
  117. [117]
    Genmab's Outlook Beyond Darzalex's Patent Cliff (NASDAQ:GMAB)
    Apr 15, 2025 · These patents, after accounting for extensions, begin to expire in 2029 (U.S.), 2030 (Japan) and 2031 (Europe). Because of the nature of the ...
  118. [118]
    DARZALEX Dominates Multiple Myeloma Market with $11.6 Billion ...
    DARZALEX (daratumumab), Janssen's CD38-targeting monoclonal antibody, has transformed multiple myeloma treatment across all lines of therapy, generating $11.6 ...
  119. [119]
    DARZALEX Continues to Redefine Multiple Myeloma Treatment with ...
    Mar 20, 2025 · DARZALEX has significant market potential as a leading monoclonal antibody for multiple myeloma treatment. Its approval in various combinations for both newly ...
  120. [120]
    JNJ report reveals $47.8B in rebates, discounts, and fees for 2024
    Jul 21, 2025 · JNJ report reveals $47.8B in rebates, discounts, and fees for 2024 ... ICYMI, 340B payments by JNJ grew from $6.0B in 2023 to $7.4B in 2024—a 23.3 ...Missing: Darzalex | Show results with:Darzalex
  121. [121]
    [PDF] Examining 340B Hospital Price Transparency, Drug Profits, and ...
    340B hospitals' own self-reported pricing data reveals that they price the top oncology drugs at 4.9 times their 340B acquisition costs, assuming a 34.7 percent.
  122. [122]
    PBM Power: The Gross-to-Net Bubble Reached $334 Billion in 2023 ...
    Jul 16, 2024 · For 2023, DCI estimates that the total value of manufacturers' gross-to-net reductions for all brand-name drugs was $334 billion.
  123. [123]
    Cost-Minimization Analysis for Subcutaneous Daratumumab in ... - NIH
    Jul 18, 2024 · In this health economic study, we aimed to evaluate the costs and consequences of a shift from intravenous daratumumab (dara-IV) to subcutaneous ...
  124. [124]
    Cost-effectiveness of adding daratumumab or bortezomib to ...
    Nov 24, 2021 · In our analysis, we found an ICER for DRd vs VRd of $1,396,318 per PFQALY and vs Rd of $530,256 per PFQALY. These results are similar to ...
  125. [125]
    Cost-effectiveness analysis of daratumumab plus bortezomib ...
    The incremental effectiveness and cost of D-VMP versus VMP were 2.21 QALYs and $501 307.6, yielding an incremental cost-effectiveness ratio (ICER) of US $226 ...
  126. [126]
    Cost-Effectiveness Analysis of Adding Daratumumab to Bortezomib ...
    Feb 28, 2021 · Results: The base case results showed that adding daratumumab to VMP provided an additional 3.00 Lys or 2.03 QALYs, at a cost of $262,526 per LY ...Abstract · Introduction · Materials and Methods · Discussion
  127. [127]
    Cost-per-responder analysis of daratumumab, bortezomib ...
    Sep 15, 2025 · Based on the PERSEUS trial, the US Food and Drug Administration (FDA) approved DVRd on July 30, 2024, for the induction and consolidation ...
  128. [128]
    A Cost-Effectiveness and Risk Analysis for Healthcare Payers
    Jun 20, 2024 · This study finds that daratumumab as a first-line treatment for myeloma exceeds the cost-effectiveness threshold considered in this evaluation.
  129. [129]
    Cost-Effectiveness of First-Line Versus Second-Line Use of ...
    Jan 7, 2021 · Daratumumab itself is priced at nearly. $6,500 in US dollars (USD) per infusion, and the total cost of treatment with regimens such as DRd can ...
  130. [130]
    Cost-effectiveness and budget impact analysis of Daratumumab ...
    Feb 28, 2024 · At the WTP threshold of $150,000, DRd was more cost-effective than VRd and Rd, with ICERs of $1,396,318 and $1,060,832 per item, respectively. ...
  131. [131]
    Losing CD38 in myeloma | Blood | American Society of Hematology
    Sep 25, 2025 · Anti-CD38 antibodies such as daratumumab and isatuximab have become foundational in myeloma therapy, from frontline therapy through each relapse ...
  132. [132]
    Monocytes and Granulocytes Reduce CD38 Expression Levels on ...
    The mechanism of CD38 reduction probably involves both elimination of CD38high multiple myeloma cells and transfer of daratumumab–CD38 complexes and ...
  133. [133]
    Resistance Mechanisms towards CD38−Directed Antibody Therapy ...
    It is important to gain insight into the mechanisms of resistance to CD38−targeting antibodies in MM, and to develop strategies to overcome this resistance.
  134. [134]
    Outcomes and prognostic indicators in daratumumab-refractory ...
    Daratumumab-refractory multiple myeloma (Dara-R MM) presents a significant treatment challenge. This study aimed to evaluate the efficacy and survival outcomes ...<|separator|>
  135. [135]
    Daratumumab Retreatment Yields Sustained Response in R/R ...
    Jun 3, 2024 · The overall response rate (ORR) in the initial daratumumab-based therapy arm (D1) was 52% vs 54% for the retreatment (D2) arm. Additional ...
  136. [136]
    Daratumumab/lenalidomide/dexamethasone in transplant-ineligible ...
    Feb 27, 2025 · In the MAIA study, daratumumab plus lenalidomide and dexamethasone (D-Rd) improved progression-free survival (PFS) and overall survival (OS) ...
  137. [137]
    Genomic and immune determinants of resistance to daratumumab ...
    Overall, this study sheds light on the intricate interplay between genomic complexity and the immune microenvironment driving resistance to Dara-Rd in patients ...
  138. [138]
    The bone marrow immune ecosystem shapes daratumumab ...
    Aug 1, 2025 · Several possible causes are described, including down-regulation of CD38 ... Mechanisms of resistance to anti-CD38 daratumumab in multiple myeloma ...
  139. [139]
    A Cost-Effectiveness and Risk Analysis for Healthcare Payers
    Jun 20, 2024 · This study finds that daratumumab as a first-line treatment for myeloma exceeds the cost-effectiveness threshold considered in this evaluation.
  140. [140]
    Cost-effectiveness and budget impact analysis of Daratumumab ...
    Feb 28, 2024 · The ICER for the DRd treatment regimen was $576,247 per QALY. One-way sensitivity analysis showed that the results were highly sensitive to the ...
  141. [141]
    Daratumumab Utilization and Cost Analysis among Patients with ...
    The estimated annual cost of mAb therapies is about US$100,000 higher in oncology and hematology than in other disease states [Citation44]. Costs of treatments ...
  142. [142]
    PCN69 COST-EFFECTIVENESS OF DARATUMUMAB ADDED TO ...
    Daratumumab therapy (DRd) had a high incremental cost effectiveness ratio (ICER) ($364,990 per progression-free life-year [PFLY]) compared to standard (Rd) care ...