MedPath

Daratumumab Advanced Drug Monograph

Published:Jul 11, 2025

Generic Name

Daratumumab

Brand Names

Darzalex, Darzalex Faspro

Drug Type

Biotech

CAS Number

945721-28-8

Associated Conditions

Multiple Myeloma (MM), Primary Amyloidosis, Refractory Multiple Myeloma, Relapsed Multiple Myeloma

Daratumumab: A Comprehensive Monograph on a First-in-Class Anti-CD38 Monoclonal Antibody

I. Introduction and Drug Profile

A. Overview of Daratumumab

Daratumumab is a first-in-class, human-specific, CD38-directed cytolytic monoclonal antibody that has fundamentally altered the treatment landscape for hematological malignancies, most notably multiple myeloma (MM) and light chain (AL) amyloidosis.[1] Classified as a protein-based therapy and antineoplastic agent, it functions by targeting the CD38 protein, which is highly and uniformly expressed on the surface of malignant plasma cells.[1] This targeted immunotherapy represents a significant departure from traditional cytotoxic chemotherapy, leveraging the patient's own immune system to eradicate cancer cells.

The medication is commercially available under two principal brand names: Darzalex®, the original formulation for intravenous (IV) infusion, and Darzalex Faspro®, a subsequent subcutaneous (SC) formulation that is co-formulated with the enzyme hyaluronidase-fihj to facilitate rapid administration.[1] The introduction of daratumumab marked a major milestone in the field, as it was the first monoclonal antibody approved to specifically target the CD38 antigen in multiple myeloma, providing a novel mechanism of action for a disease characterized by recurrent relapse and eventual resistance to other therapies.[5]

B. Development and Commercialization

The journey of daratumumab from laboratory discovery to a cornerstone of clinical practice began with its original development by the biotechnology company Genmab A/S.[2] A pivotal moment in its trajectory occurred in August 2012, when Genmab entered into a worldwide agreement with Janssen Biotech, Inc., a pharmaceutical company of Johnson & Johnson. This agreement granted Janssen an exclusive license to develop, manufacture, and commercialize the drug globally.[2] This strategic partnership provided the substantial resources necessary to conduct the large-scale, pivotal clinical trials that would ultimately lead to its widespread regulatory approval and integration into standard-of-care regimens.[1] Throughout its development and in clinical literature, daratumumab is also referred to by its development codes, including HuMax-CD38 and JNJ-54767414.[1]

Table 1: Key Identifiers and Physicochemical Properties of Daratumumab

This table consolidates the fundamental identifiers and structural properties of the molecule, serving as a foundational reference for researchers, clinicians, and regulatory experts.

PropertyValueSource(s)
Generic NameDaratumumab1
Brand NamesDarzalex, Darzalex Faspro1
DrugBank IDDB093311
CAS Number945721-28-81
TypeBiotech, Monoclonal Antibody (mAb)1
SourceHuman2
IsotypeHuman Immunoglobulin G1 kappa (IgG1κ)1
Molecular FormulaC6466​H9996​N1724​O2010​S42​1
Average Molecular WeightApprox. 145,391.67 Da (~148 kDa)1
ATC CodeL01FC012
UNII4Z63YK6E0E2
KEGG IDD1077713

II. Molecular Structure and Manufacturing

A. Molecular Structure

Daratumumab is a fully human monoclonal antibody of the immunoglobulin G1 kappa (IgG1κ) subclass.[3] Its structure is characteristic of a typical antibody, comprising two identical heavy chains and two identical light chains. These polypeptide chains are linked covalently by a series of disulfide bonds, forming a symmetric, Y-shaped dimeric molecule with a molecular weight of approximately 148 kDa.[4]

The precise amino acid sequences of the heavy and light chains define its specificity and effector functions.

  • Heavy Chain: The heavy chain consists of 452 amino acids with the sequence: EVQLLESGGGLVQPGGSLRLSCAVSGFTFNSFAMSWVRQAPGKGLEWVSAISGSGGGTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYFCAKDKILWFGEPVFDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK.[1]
  • Light Chain: The light chain is composed of 214 amino acids with the sequence: EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLLIYDASNRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSNWPPTFGQGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC.[1]

The three-dimensional structure of the antibody is stabilized by a network of 16 disulfide bridges. These include intra-chain bonds that form the characteristic immunoglobulin domains (e.g., H22-H96) and inter-chain bonds that link the heavy and light chains (e.g., H225-L214) and the two heavy chains together in the flexible hinge region (e.g., H231-H'231).[13] The variable regions of the heavy and light chains combine to form the antigen-binding site (Fab fragment), which recognizes and binds with high affinity to a unique conformational epitope on the CD38 protein. This binding site is distinct from that of isatuximab, another clinically approved anti-CD38 antibody, which may account for subtle differences in their biological activities.[2]

B. Manufacturing Process

The production of daratumumab is a complex, multi-stage biopharmaceutical process that relies on recombinant DNA technology to ensure a consistent and high-purity product.[12] The process begins with a well-characterized cell line and is broadly divided into upstream and downstream operations.

  • Cell Line and Upstream Processing: Daratumumab is produced in a mammalian cell line, specifically Chinese Hamster Ovary (CHO) cells, which are capable of performing the complex protein folding and post-translational modifications (e.g., glycosylation) required for a functional human antibody.[12] The manufacturing process starts with the thawing of a vial from a validated working cell bank (WCB), which itself is derived from a master cell bank (MCB) to ensure genetic consistency across all production batches.[21] The cells are expanded through a seed train into large-scale bioreactors. Here, they are grown in a nutrient-rich culture medium under tightly controlled conditions (e.g., pH, temperature, dissolved oxygen) in a fed-batch system, where nutrients are added periodically to maximize cell density and antibody production over the culture period.[20]
  • Downstream Processing and Formulation: Once the cell culture phase is complete, the antibody must be harvested and purified. This downstream process is critical for removing the host cells, cellular debris, and other process-related impurities. The active substance is manufactured in what has been described as an 11-stage process.[22] Key steps include:
  1. Harvest and Clarification: The bioreactor contents are first clarified to separate the antibody-containing supernatant from the CHO cells and debris, typically using centrifugation and/or depth filtration.[20]
  2. Purification: The clarified supernatant undergoes a series of chromatography steps. This usually begins with Protein A affinity chromatography, which specifically binds the Fc region of the IgG antibody, providing a highly effective initial purification step. This is followed by additional polishing steps, such as ion-exchange and hydrophobic interaction chromatography, to remove remaining impurities like host cell proteins and DNA.[20]
  3. Viral Safety: The process includes dedicated viral inactivation (e.g., low pH treatment) and viral filtration steps to ensure the final product is free from potential viral contaminants.[22]
  4. Formulation and Final Fill: The highly purified daratumumab is concentrated and formulated into its final buffer solution, which contains excipients such as sorbitol, L-histidine, and polysorbate 20 to maintain stability.[18] The final drug product is then sterile-filtered and aseptically filled into single-dose glass vials, such as 100 mg/5 mL and 400 mg/20 mL presentations for the intravenous formulation.[12]

The intricate and resource-intensive nature of this manufacturing process is not merely a technical footnote; it directly influences the drug's clinical and logistical characteristics. The initial intravenous formulation, a direct output of this process, required lengthy infusion times—often up to seven hours for the first dose—and had to be administered in a specialized clinical setting equipped to manage potentially severe infusion-related reactions.[6] This created a significant logistical and time burden for both patients and healthcare systems. The development of the subcutaneous formulation, Darzalex Faspro, was a direct and innovative response to these challenges. By co-formulating daratumumab with the recombinant human enzyme hyaluronidase, the drug product can be administered in approximately 3-5 minutes.[1] The hyaluronidase temporarily and locally degrades hyaluronic acid in the subcutaneous space, increasing tissue permeability and allowing for the rapid dispersion and absorption of a large volume of the antibody that would otherwise be impossible to deliver via this route.[7] This evolution from an IV to an SC formulation exemplifies a critical trend in biopharmaceutical development, where initial proof of efficacy is followed by subsequent innovation focused on enhancing patient convenience, improving safety profiles, and reducing the burden on healthcare infrastructure.

III. Comprehensive Pharmacology

A. Mechanism of Action

The therapeutic efficacy of daratumumab is predicated on its high-affinity binding to the CD38 glycoprotein, a target that defines its mechanism of action.[2] CD38 is a multifunctional type II transmembrane protein that functions as both a receptor involved in cell adhesion and a key enzyme (ADP-ribosyl cyclase) in nucleotide metabolism.[1] Its expression pattern is the key to daratumumab's therapeutic window. CD38 is highly and uniformly expressed on the surface of malignant plasma cells in both multiple myeloma and AL amyloidosis. While it is also present on some normal hematopoietic cells—including natural killer (NK) cells, T cells, B cells, and red blood cells—the level of expression is significantly lower than on their malignant counterparts.[2] This differential expression allows daratumumab to preferentially target and eliminate the cancerous cell population.

Upon binding to CD38, daratumumab unleashes a multi-pronged attack on the tumor cell, mediated primarily by the Fc domain of its IgG1 structure. This induces cell death through several distinct, immune-mediated pathways [2]:

  • Complement-Dependent Cytotoxicity (CDC): The Fc region of daratumumab binds to the C1q component of the complement system, initiating the classical complement cascade. This enzymatic cascade culminates in the formation of the membrane attack complex (MAC), a pore-like structure that inserts into the myeloma cell membrane, leading to osmotic lysis and rapid cell death.[11] Daratumumab is recognized as a particularly potent inducer of CDC, a mechanism that distinguishes it from some other anti-CD38 antibodies.[11]
  • Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC): The daratumumab Fc domain also engages Fc gamma receptors (FcγR) on the surface of immune effector cells, most notably NK cells. This engagement activates the NK cells, causing them to release cytotoxic granules containing perforin and granzymes, which induce apoptosis in the targeted myeloma cell.[2]
  • Antibody-Dependent Cellular Phagocytosis (ADCP): In a similar Fc-mediated process, daratumumab-coated (opsonized) myeloma cells are recognized by FcγRs on phagocytic cells, primarily macrophages. This triggers the engulfment and destruction of the cancer cell by the macrophage.[2] This mechanism is thought to be particularly important within the protective bone marrow microenvironment where macrophages are abundant.[11]
  • Induction of Apoptosis: In addition to immune-mediated killing, the cross-linking of CD38 molecules on the tumor cell surface by daratumumab binding can directly trigger intracellular signaling pathways that lead to programmed cell death, or apoptosis.[2]

Beyond these direct cytotoxic effects, daratumumab exerts powerful immunomodulatory functions by reshaping the tumor microenvironment. It eliminates CD38-expressing immunosuppressive cell populations that protect the tumor from immune surveillance.[15] This includes the depletion of regulatory T cells (CD38+ Tregs), regulatory B cells (CD38+ Bregs), and myeloid-derived suppressor cells (CD38+ MDSCs).[15] The removal of these suppressive elements leads to a more favorable immune milieu, characterized by an increased ratio of cytotoxic CD8+ T cells to helper CD4+ T cells and enhanced T-cell clonal expansion, thereby augmenting the patient's intrinsic anti-tumor immune response.[28]

B. Pharmacokinetics (PK)

The pharmacokinetic profile of daratumumab describes how the body absorbs, distributes, metabolizes, and eliminates the drug. This profile is characterized by nonlinear kinetics, primarily due to its mechanism of action.

  • Absorption and Bioavailability:
  • Intravenous (IV) Formulation (Darzalex): When administered intravenously, daratumumab has 100% bioavailability as it is delivered directly into the systemic circulation. Pharmacokinetic studies show that the maximum concentration (Cmax​) is generally dose-proportional, whereas the area under the concentration-time curve (AUC) increases more than proportionally with the dose. This is a classic feature of drugs with saturable, target-mediated clearance.[31]
  • Subcutaneous (SC) Formulation (Darzalex Faspro): The co-formulation with recombinant human hyaluronidase is essential for its absorption. The hyaluronidase temporarily degrades hyaluronan in the subcutaneous matrix, increasing its permeability and allowing the large volume of the 1,800 mg flat dose to be rapidly absorbed.[1] The pivotal Phase 3 COLUMBA trial (NCT03277105) established the non-inferiority of the SC formulation to the standard 16 mg/kg IV dose. The study met its co-primary endpoints, demonstrating a comparable overall response rate (ORR) of 41.1% for SC versus 37.1% for IV, and achieving non-inferior C-trough concentrations on Cycle 3, Day 1.[17] In fact, long-term follow-up showed that the SC formulation can result in comparable or even slightly higher trough concentrations over time compared to the IV formulation.[17]
  • Distribution: Daratumumab has a small volume of distribution, with the central compartment volume estimated to be between 4 and 5 liters.[1] This indicates that the drug is primarily localized within the vascular system, with limited distribution into extravascular tissues, a typical characteristic for large monoclonal antibodies.[1]
  • Metabolism: As a large protein, daratumumab is not metabolized by the hepatic cytochrome P450 enzyme system that is responsible for clearing most small-molecule drugs. Instead, it is presumed to undergo catabolism, where it is broken down into smaller peptides and amino acids by proteolytic enzymes throughout the body. These components are then recycled for de novo protein synthesis or eliminated.[1]
  • Elimination and Clearance: The elimination of daratumumab is complex and nonlinear, a direct consequence of its high-affinity binding to the CD38 target. This phenomenon, known as target-mediated drug disposition (TMDD), results in a clearance rate that is both concentration- and time-dependent.[15]
  • At the beginning of therapy, when CD38 expression on myeloma cells is high, a significant portion of the drug is cleared rapidly by binding to its target.
  • As treatment continues and the tumor burden decreases, the CD38 target becomes saturated. This saturable clearance pathway is overwhelmed, leading to a decrease in the overall clearance rate and a corresponding increase in the drug's half-life.
  • After repeated dosing with the 16 mg/kg IV regimen, the terminal elimination half-life is approximately 18 days.[1] The SC formulation has a similarly long half-life of around 20 days, which supports the less frequent dosing schedules (every two to four weeks) used in the maintenance phase of treatment.[1]
  • Importantly, clearance is not significantly impacted by patient factors such as mild hepatic impairment or renal impairment, meaning dose adjustments are generally not required for these conditions.[15]

C. Pharmacodynamics (PD)

The pharmacodynamic effects of daratumumab encompass its direct impact on the body, including the desired therapeutic response and its notable interferences with common laboratory tests.

  • Exposure-Response Relationship: There is a clear relationship between daratumumab exposure and clinical efficacy. Analyses from seminal trials demonstrated that the overall response rate increases with rising daratumumab concentrations, reaching a plateau at trough levels that ensure near-complete (>99%) saturation of the CD38 target on myeloma cells.[15] The approved dosing regimens—16 mg/kg for IV and a flat dose of 1,800 mg for SC—were specifically chosen to achieve and maintain these saturating concentrations throughout the dosing intervals, thereby maximizing the drug's therapeutic potential.[15] Conversely, no significant exposure-safety relationship has been identified for key adverse events like infusion reactions or cytopenias, suggesting these are not strictly dose-dependent within the therapeutic range.[34]
  • Laboratory Test Interferences: Daratumumab's on-target binding to CD38 on non-malignant cells, coupled with its nature as a therapeutic antibody, leads to predictable and clinically significant interferences with standard laboratory tests.
  • Interference with Serological Testing (Blood Bank): Daratumumab binds to the low levels of CD38 present on the surface of red blood cells (RBCs). This in-vivo coating of RBCs causes a pan-reactive positive result in the indirect antiglobulin test (also known as the Indirect Coombs test), which is a cornerstone of pre-transfusion compatibility testing. This interference can persist for up to six months after the last dose of daratumumab.[2] The consequence is that the test cannot reliably detect the presence of clinically significant alloantibodies in the patient's plasma, which could lead to a delayed or hemolytic transfusion reaction if incompatible blood is given.
  • Clinical Management: To ensure patient safety, a strict protocol must be followed. A baseline blood type and antibody screen (including Rh and Kell phenotyping) must be performed before the patient receives their first dose of daratumumab.[2] The blood transfusion center must be formally notified that the patient is receiving daratumumab. To perform subsequent cross-matches, transfusion labs can use mitigation techniques, most commonly treating reagent RBCs with dithiothreitol (DTT). DTT cleaves the disulfide bonds on CD38, removing the daratumumab interference. However, DTT also destroys antigens in the Kell system, necessitating the provision of Kell-negative blood units unless alloantibodies can be definitively ruled out.[2]
  • Interference with M-Protein Monitoring: As a therapeutic human IgG kappa monoclonal antibody, daratumumab itself can be detected by the laboratory assays used to monitor the patient's own monoclonal protein (M-protein)—namely, serum protein electrophoresis (SPE) and immunofixation (IFE).[19] This creates a significant challenge in assessing disease response, as the therapeutic antibody can migrate to the same position as the patient's native M-protein, leading to a false-positive result. This interference can make it difficult to accurately determine if a patient has achieved a true complete response (CR), where the endogenous M-protein is absent.
  • Clinical Management: To overcome this, a daratumumab-specific immunofixation electrophoresis (D-IFE) assay has been developed and validated. This specialized test can distinguish the daratumumab protein from the patient's endogenous M-protein, allowing for an accurate assessment of deep responses in patients with IgG kappa myeloma.[25]

These pharmacodynamic characteristics create a unique "clinical footprint" for daratumumab, extending beyond typical drug toxicities. The on-target effects on non-malignant cells (RBCs) and its physical properties as a therapeutic protein necessitate a proactive, multidisciplinary management strategy. Safe and effective administration requires seamless coordination between the hematology-oncology team, the blood bank, and the clinical pathology laboratory. Before the first dose is even administered, the oncologist must anticipate these interferences by ordering a baseline type and screen, notifying the blood bank, being aware of the potential for SPE/IFE interference, and knowing when to order a D-IFE assay. This level of system-wide protocol and inter-departmental communication is a direct consequence of the drug's fundamental pharmacology and is essential for ensuring both patient safety during transfusions and the accurate monitoring of their response to therapy.

IV. Regulatory History and Global Approvals

The regulatory journey of daratumumab has been characterized by a rapid and expansive series of approvals from major health authorities, reflecting its profound clinical impact. Its trajectory showcases a clear path from a therapy for heavily pretreated patients to a cornerstone of frontline treatment.

A. U.S. Food and Drug Administration (FDA) Trajectory

The FDA's review and approval process for daratumumab has been notably swift, facilitated by multiple expedited programs.

  • Early Designations and Initial Approval: Daratumumab was granted Breakthrough Therapy Designation in May 2013 for the treatment of patients with multiple myeloma who had received at least two prior therapies, including a proteasome inhibitor (PI) and an immunomodulatory agent (IMiD), and were refractory to both.[2] This designation, along with an Orphan Drug status for MM, underscored its potential to address a significant unmet need.[2] On November 16, 2015, the FDA granted accelerated approval for daratumumab as a monotherapy for patients with RRMM who had received at least three prior lines of therapy. This initial approval was based on the compelling overall response rate observed in the Phase 2 SIRIUS study.[1]
  • Expansion into Combination Therapy and Earlier Lines (2016-2019): Following its monotherapy approval, daratumumab's indications rapidly expanded as pivotal Phase 3 trials demonstrated its efficacy as a combination partner.
  • In November 2016, it was approved in combination with either lenalidomide and dexamethasone (Rd) or bortezomib and dexamethasone (Vd) for patients with MM who had received at least one prior therapy, based on the groundbreaking results of the POLLUX and CASTOR trials, respectively.[2]
  • Subsequent approvals included its use with pomalidomide and dexamethasone (Pd) in June 2017 for patients with at least two prior therapies.[5]
  • A critical milestone was reached in May 2018 with its first approval in the frontline setting for newly diagnosed multiple myeloma (NDMM). Based on the ALCYONE study, it was approved in combination with bortezomib, melphalan, and prednisone (VMP) for patients ineligible for autologous stem cell transplant (ASCT).[2]
  • In September 2019, its use was extended to transplant-eligible NDMM patients in combination with bortezomib, thalidomide, and dexamethasone (VTd).[36]
  • Recent Approvals of New Formulations and Quadruplet Regimens (2020-2024):
  • On May 1, 2020, the FDA approved the subcutaneous formulation, Darzalex Faspro (daratumumab and hyaluronidase-fihj). This was a major advancement, offering comparable efficacy with significantly improved patient convenience and a better safety profile regarding administration reactions.[2]
  • On January 15, 2021, daratumumab received accelerated approval for a new disease indication: newly diagnosed systemic AL amyloidosis, in combination with bortezomib, cyclophosphamide, and dexamethasone (VCd), based on the ANDROMEDA trial.[7]
  • On July 30, 2024, the FDA approved the landmark quadruplet regimen of Darzalex Faspro combined with bortezomib, lenalidomide, and dexamethasone (D-VRd) for the induction and consolidation treatment of transplant-eligible NDMM patients. This approval was based on the superior efficacy demonstrated in the PERSEUS trial.[1]

B. European Medicines Agency (EMA) Trajectory

The EMA's approval pathway for daratumumab has largely paralleled that of the FDA, establishing it as a standard of care across Europe.

  • Initial Authorization and Extensions: Daratumumab first received a conditional marketing authorization from the EMA on May 20, 2016, as a monotherapy for RRMM.[2] This was subsequently converted to a full, standard authorization in April 2017 as more data became available.[42] Over the following years, the EMA progressively expanded the approved indications to include the same key combinations as the FDA for both RRMM and NDMM (transplant-eligible and -ineligible), as well as for AL amyloidosis.[2]
  • Recent and Forthcoming Approvals: The EMA has continued to broaden the scope of daratumumab's use based on the latest clinical evidence.
  • Following a positive opinion from the Committee for Medicinal Products for Human Use (CHMP) in October 2024, the European Commission officially approved the D-VRd regimen for transplant-eligible NDMM patients based on the PERSEUS study results.[45]
  • In April 2025, the European Commission further extended this approval, authorizing the D-VRd regimen for all adult patients with NDMM, regardless of their transplant eligibility. This broader approval was supported by data from both the PERSEUS and CEPHEUS trials, making the quadruplet regimen a standard of care for the entire frontline NDMM population in the EU.[47]
  • In a significant step towards early intervention, the CHMP issued a positive opinion in June 2025 recommending a new indication for daratumumab monotherapy for the treatment of adult patients with high-risk smoldering multiple myeloma (SMM), potentially making it the first approved therapy for this precursor condition.[44]

Table 2: Summary of Key Approved Indications for Daratumumab (FDA & EMA)

The regulatory landscape for daratumumab is exceptionally complex. This table provides a structured overview to clarify the approved use cases, specifying patient populations, combination regimens, and the responsible regulatory body.

Patient PopulationLine of TherapyApproved RegimenPivotal Trial(s)FDA ApprovalEMA Approval
Newly Diagnosed MM (NDMM), Transplant-EligibleFrontlineDaratumumab + Bortezomib, Lenalidomide, Dexamethasone (D-VRd)PERSEUSYesYes
FrontlineDaratumumab + Bortezomib, Thalidomide, Dexamethasone (D-VTd)CASSIOPEIAYesYes
Newly Diagnosed MM (NDMM), Transplant-IneligibleFrontlineDaratumumab + Lenalidomide, Dexamethasone (D-Rd)MAIAYesYes
FrontlineDaratumumab + Bortezomib, Melphalan, Prednisone (D-VMP)ALCYONEYesYes
FrontlineDaratumumab + Bortezomib, Lenalidomide, Dexamethasone (D-VRd)CEPHEUSNoYes
Relapsed/Refractory MM (RRMM)≥1 Prior LineDaratumumab + Lenalidomide, Dexamethasone (D-Rd)POLLUXYesYes
≥1 Prior LineDaratumumab + Bortezomib, Dexamethasone (D-Vd)CASTORYesYes
≥1-3 Prior LinesDaratumumab + Carfilzomib, Dexamethasone (D-Kd)CANDORYesYes
≥2 Prior LinesDaratumumab + Pomalidomide, Dexamethasone (D-Pd)EQUULEUSYesYes
≥3 Prior LinesDaratumumab MonotherapySIRIUSYesYes
Newly Diagnosed Light Chain (AL) AmyloidosisFrontlineDaratumumab + Bortezomib, Cyclophosphamide, Dexamethasone (D-VCd)ANDROMEDAYes (Accelerated)Yes
High-Risk Smoldering MM (SMM)Pre-MalignantDaratumumab MonotherapyAQUILANoPositive Opinion

V. Clinical Efficacy in Multiple Myeloma and AL Amyloidosis

The clinical development program for daratumumab has been extensive, with numerous pivotal Phase 3 trials consistently demonstrating its ability to improve outcomes when added to existing standards of care across the entire spectrum of multiple myeloma and in AL amyloidosis.

A. Newly Diagnosed Multiple Myeloma (NDMM)

The greatest impact of daratumumab has arguably been in the frontline setting, where it has redefined treatment goals for both transplant-eligible and transplant-ineligible patients.

  • Transplant-Eligible Patients:
  • The PERSEUS trial (NCT03710603) is a landmark study that established the superiority of quadruplet therapy in this population. It was a randomized, open-label trial that compared the subcutaneous daratumumab-based regimen of D-VRd (daratumumab, bortezomib, lenalidomide, dexamethasone) to the standard triplet VRd.[40] The results were unequivocal. At a median follow-up of nearly four years, the addition of daratumumab resulted in a striking 58% reduction in the risk of disease progression or death, with a hazard ratio (HR) for progression-free survival (PFS) of 0.42.[50] The 4-year PFS rate was 84% in the D-VRd arm compared to 68% in the VRd arm.[50] This benefit was driven by a much deeper response, with significantly higher rates of minimal residual disease (MRD) negativity at a sensitivity of 10−5 (75% for D-VRd vs. 48% for VRd).[40] Critically, these deep responses were durable, with 65% of patients in the daratumumab arm achieving sustained MRD negativity for at least 12 months, compared to only 30% in the control arm.[50]
  • The earlier CASSIOPEIA trial (MMY3006) evaluated the addition of intravenous daratumumab to the VTd (bortezomib, thalidomide, dexamethasone) backbone. It also demonstrated a significant improvement in the depth of response, with a higher rate of stringent complete response (sCR) post-consolidation in the daratumumab arm (29% vs. 20%), providing early evidence for the benefit of quadruplet induction.[42]
  • Transplant-Ineligible Patients:
  • The MAIA trial (MMY3008) randomized patients to receive either daratumumab plus lenalidomide and dexamethasone (D-Rd) or Rd alone.[42] The addition of daratumumab led to a significant improvement in PFS, with a 44% reduction in the risk of progression or death (HR 0.56) and a much higher overall response rate (93% vs. 81%).[42]
  • Similarly, the ALCYONE trial (MMY3007) compared daratumumab plus bortezomib, melphalan, and prednisone (D-VMP) to VMP alone.[5] The D-VMP regimen resulted in a 50% reduction in the risk of progression or death (HR 0.50) and a markedly higher ORR (91% vs. 74%).[5]
  • The more recent CEPHEUS trial (NCT03652064) investigated the potent D-VRd quadruplet regimen versus VRd in transplant-ineligible or transplant-deferred patients.[47] This trial confirmed the benefit of the four-drug combination in this population, showing significantly higher rates of MRD negativity (60.9% vs. 39.4%) and a substantial improvement in PFS (median not reached vs. 52.6 months; HR 0.57).[47]

B. Relapsed/Refractory Multiple Myeloma (RRMM)

Daratumumab's initial approvals were in the relapsed/refractory setting, where it also demonstrated profound efficacy.

  • The POLLUX trial (NCT02076009) evaluated D-Rd versus Rd in patients with at least one prior line of therapy.[30] The combination significantly improved PFS and nearly doubled the rate of complete response or better, establishing D-Rd as a standard of care in early relapse.[52]
  • The CASTOR trial (NCT02136134) compared D-Vd versus Vd in a similar patient population.[30] Again, the addition of daratumumab led to a significant PFS benefit and higher response rates, providing another powerful combination option for relapsed patients.[52]
  • The SIRIUS trial, which supported the drug's initial accelerated approval, demonstrated meaningful single-agent activity in a heavily pretreated population (median of 5 prior lines of therapy). It achieved an ORR of 29% with a median duration of response of 7.4 months, providing a crucial new option for patients who had exhausted other therapies.[33]

C. Light Chain (AL) Amyloidosis

The efficacy of daratumumab extends beyond multiple myeloma to the related plasma cell disorder, AL amyloidosis.

  • The pivotal ANDROMEDA trial (NCT03201965) was a Phase 3 study that randomized newly diagnosed patients to receive either subcutaneous daratumumab plus the standard backbone of bortezomib, cyclophosphamide, and dexamethasone (D-VCd) or VCd alone.[39] The trial met its primary endpoint, showing that the daratumumab-containing regimen led to a significantly higher rate of hematologic complete response compared to the control arm. These results led to its accelerated approval for this indication, offering the first approved targeted therapy for this rare and devastating disease.[39]

D. The Emergence of Minimal Residual Disease (MRD) as a Key Endpoint

The clinical development of daratumumab has coincided with, and indeed helped to propel, a major shift in how therapeutic success is measured in multiple myeloma. The focus has decisively moved beyond traditional serologic and imaging-based response criteria (e.g., CR, VGPR) toward the goal of achieving deep, molecular remission, as measured by minimal residual disease (MRD) negativity. This evolution is not merely a technical one; it reflects a fundamental change in treatment philosophy, from disease control to curative intent.

The initial daratumumab trials in the relapsed setting, such as SIRIUS, POLLUX, and CASTOR, primarily used ORR and PFS as their key endpoints.[34] However, as the drug moved into the frontline setting, its profound efficacy necessitated more sensitive tools to differentiate the depth of response. The recent landmark trials—PERSEUS, CEPHEUS, and ADVANCE—have all prominently featured MRD negativity as a critical endpoint.[40] The unprecedented rates of MRD negativity achieved with daratumumab-based quadruplet regimens (e.g., 75% in the PERSEUS D-VRd arm) are directly correlated with their superior PFS outcomes.[40] This strong and consistent correlation has solidified the prognostic power of MRD; achieving a sustained MRD-negative state is now widely accepted as the strongest predictor of long-term PFS and, ultimately, overall survival.[40]

This paradigm shift has profound implications for both clinical practice and future research. The FDA's Oncologic Drugs Advisory Committee has voted in favor of using MRD as a surrogate endpoint for accelerated approval in myeloma trials, a move that will significantly shorten drug development timelines.[59] For patients, this opens the door to MRD-guided therapy. Clinical trials are already underway to determine whether patients who achieve a deep, sustained MRD-negative response can safely de-escalate or even discontinue maintenance therapy.[40] This personalized approach, guided by the depth of response that daratumumab helps to achieve, holds the promise of reducing the long-term toxicity, financial burden, and quality-of-life impact of continuous treatment, representing the next frontier in myeloma care.

Table 3: Pivotal Phase 3 Clinical Trial Summary

This table synthesizes the vast amount of clinical trial data into a structured, comparable format, allowing for a quick understanding of the evidence supporting each key indication.

Trial Name (NCT ID)Patient PopulationTreatment ArmsPrimary EndpointKey Efficacy Outcome(s)
PERSEUS (NCT03710603)NDMM, Transplant-EligibleD-VRd vs. VRdPFSPFS HR: 0.42; 4-yr PFS: 84% vs. 68%; MRD-negativity: 75% vs. 48%
CEPHEUS (NCT03652064)NDMM, Transplant-IneligibleD-VRd vs. VRdMRD-Negativity RateMRD-negativity: 60.9% vs. 39.4%; PFS HR: 0.57
MAIA (NCT02579181)NDMM, Transplant-IneligibleD-Rd vs. RdPFSPFS HR: 0.56; ORR: 93% vs. 81%
ALCYONE (NCT02195479)NDMM, Transplant-IneligibleD-VMP vs. VMPPFSPFS HR: 0.50; ORR: 91% vs. 74%
POLLUX (NCT02076009)RRMM (≥1 prior line)D-Rd vs. RdPFSPFS HR: 0.37; ORR: 93% vs. 76%
CASTOR (NCT02136134)RRMM (≥1 prior line)D-Vd vs. VdPFSPFS HR: 0.33; ORR: 83% vs. 63%
ANDROMEDA (NCT03201965)Newly Diagnosed AL AmyloidosisD-VCd vs. VCdHematologic CR RateSignificantly higher hema-CR rate with D-VCd (p<0.0001)

VI. Comprehensive Safety and Tolerability Profile

The safety profile of daratumumab is well-characterized across numerous clinical trials involving thousands of patients. While generally considered manageable, it includes a distinct set of adverse events related to its on-target mechanism of action, requiring specific monitoring and management protocols.

A. Overview of Adverse Events (AEs)

The most frequently reported adverse reactions (occurring in ≥20% of patients) across various studies and combination regimens are infusion-related reactions (with the IV formulation), fatigue, nausea, diarrhea, constipation, pyrexia, cough, dyspnea, and upper respiratory tract infections.[3] Hematologic toxicities are also very common and are often exacerbated when daratumumab is added to other myelosuppressive agents. These include neutropenia, thrombocytopenia, and anemia.[3]

B. Infusion-Related Reactions (IRRs) and Hypersensitivity

Infusion-related reactions are the most characteristic and common adverse event associated with the intravenous formulation of daratumumab.

  • Incidence and Timing: IRRs are reported in approximately 40-50% of patients receiving IV daratumumab. A crucial feature is that the vast majority of these reactions—between 80% and 95%—occur during the very first infusion.[15] The incidence decreases dramatically with subsequent infusions, falling to just a few percent.[32] The median time to onset is about 1.5 hours after the start of the infusion.[32]
  • Clinical Presentation: Most IRRs are Grade 1 or 2 in severity. Common symptoms include nasal congestion, chills, cough, throat irritation, dyspnea, and nausea.[18] However, severe (Grade 3/4) reactions, including anaphylaxis, bronchospasm, hypoxia, and laryngeal edema, can occur, although they are rare. These reactions can be life-threatening, and fatal outcomes have been reported.[3]
  • Mandatory Management Protocol: Due to the high incidence and potential severity of IRRs, a strict management protocol is required for all patients.
  1. Pre-medication: All patients must receive pre-medication 1-3 hours before every infusion or injection. This consists of a corticosteroid (e.g., methylprednisolone or dexamethasone), an antipyretic (acetaminophen), and an H1-receptor antagonist (e.g., diphenhydramine).[3]
  2. Infusion Management: Patients must be closely monitored during the infusion. If a reaction of any severity occurs, the infusion must be interrupted immediately, and supportive medical management instituted. For Grade 1-3 reactions, the infusion may be resumed at a reduced rate once symptoms have resolved. For a Grade 4 (life-threatening) reaction, daratumumab must be permanently discontinued.[18]
  3. Post-medication: To mitigate the risk of delayed reactions, patients are prescribed an oral corticosteroid for 1-2 days following the infusion.[18]
  • Advantage of Subcutaneous Formulation: A major clinical advantage of the subcutaneous formulation (Darzalex Faspro) is a significantly lower rate of administration-related reactions compared to the IV formulation, making it a safer and more convenient option for many patients.[15]

C. Hematologic Toxicity and Infections

  • Myelosuppression: Daratumumab can exacerbate the myelosuppressive effects of its combination partners. Rates of Grade 3/4 neutropenia and thrombocytopenia are consistently higher in daratumumab-containing arms compared to control arms in clinical trials.[25] Therefore, regular monitoring of complete blood counts is essential throughout treatment, and dose delays of the background therapy may be required to allow for hematopoietic recovery.[26]
  • Infections: An increased risk of infection is a significant concern, stemming from a combination of the underlying immune dysfunction in myeloma and the on-target effects of daratumumab, which depletes normal CD38-expressing immune cells like B-cells and NK cells.[2]
  • Upper respiratory tract infections and pneumonia are the most frequently reported infections, and pneumonia is among the most common serious adverse events.[18]
  • Herpes Zoster Reactivation: There is a well-established risk of reactivating the latent varicella-zoster virus (shingles). To prevent this, antiviral prophylaxis (e.g., with acyclovir or valacyclovir) is recommended for all patients. Prophylaxis should be initiated within one week of starting daratumumab and continued for at least three months after the completion of treatment.[4]
  • Hepatitis B Virus (HBV) Reactivation: Although rare, cases of HBV reactivation, including fatal outcomes, have been reported. It is recommended that all patients undergo HBV screening prior to initiating therapy.[8]

D. Contraindications and Other Key Warnings

  • Contraindications: Daratumumab is contraindicated in patients with a history of a severe hypersensitivity reaction (e.g., anaphylaxis) to daratumumab or any of its excipients.[18]
  • Embryo-Fetal Toxicity: As an IgG1 antibody, daratumumab is known to cross the placental barrier. Based on its mechanism of action, it can cause fetal harm, including the depletion of fetal immune cells and potentially decreased bone density. Women of childbearing potential must be advised of this risk and should use effective contraception during treatment and for three months after the final dose.[25]
  • Cardiac Toxicity in AL Amyloidosis: A specific warning applies to the use of daratumumab in patients with AL amyloidosis, who often have significant underlying cardiac dysfunction. Serious and fatal cardiac adverse events have occurred in this population. Therefore, its use is not recommended in patients with advanced cardiac disease (NYHA Class IIIB or IV, or Mayo Stage IIIB) outside the context of a controlled clinical trial.[7]
  • Hepatotoxicity: While mild and transient elevations in liver enzymes have been observed in clinical trials, daratumumab has not been linked to cases of clinically apparent liver injury with jaundice. The National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) assigns it a hepatotoxicity likelihood score of E, indicating it is an "unlikely cause of clinically apparent liver injury".[3]

Table 4: Incidence of Key Grade ≥3 Adverse Events Across Major Clinical Trials

This table provides a quantitative, comparative view of the safety profile of daratumumab when added to different standard-of-care backbones, helping clinicians understand the incremental toxicity.

Adverse EventD-Rd (POLLUX)Rd (POLLUX)D-VMP (ALCYONE)VMP (ALCYONE)D-VRd (PERSEUS)VRd (PERSEUS)
Neutropenia51.9%37.0%39.9%38.7%44.2%29.7%
Thrombocytopenia2.5%3.2%34.4%37.6%28.4%20.0%
Anemia14.4%19.6%15.9%19.8%13.2%11.8%
Pneumonia12.3%10.1%11.3%4.0%11.2%*4.6%*
Sepsis3.2%2.5%2.3%1.1%N/AN/A
Any Infection32.1%28.4%23.1%14.7%57%**49%**

Note: Data for PERSEUS reflects COVID-19 infection rates, a prominent infection during the trial period. [47]

*Note: Data for PERSEUS reflects Serious Adverse Events of any kind, with infection being a major contributor. 50

VII. Evolving Treatment Paradigms and Future Directions

The integration of daratumumab into clinical practice has not only improved patient outcomes but has also catalyzed a rapid evolution in treatment strategies and spurred new avenues of research aimed at further optimizing therapy for multiple myeloma.

A. Comparative Analysis: Daratumumab vs. Isatuximab

The approval of a second anti-CD38 antibody, isatuximab, has introduced a new dynamic into the treatment landscape.

  • Mechanism and Preclinical Differences: Both daratumumab and isatuximab are human IgG1κ monoclonal antibodies that target CD38. However, they bind to distinct, non-overlapping epitopes on the CD38 protein, which may underlie some subtle differences in their mechanisms of action.[2] Preclinical studies suggest that while both are effective, daratumumab is a more potent inducer of complement-dependent cytotoxicity (CDC), whereas isatuximab may exhibit stronger direct apoptotic activity without the need for Fc-receptor cross-linking and a more pronounced inhibition of CD38's ecto-enzymatic function.[2]
  • Clinical Efficacy and Interchangeability: In the clinical setting, both antibodies have demonstrated profound efficacy when combined with standard-of-care backbones like proteasome inhibitors and IMiDs in both newly diagnosed and relapsed/refractory patients.[62] In the absence of direct head-to-head comparative trials, most experts consider the two agents to be clinically equivalent and largely interchangeable for the majority of patients.[65] The choice between them is often driven by practical considerations, such as the specific combination regimen being used (as different pivotal trials have supported different combinations, e.g., PERSEUS for D-VRd, IsKia for Isa-KRd), regional approvals, and logistical factors like dosing schedules and infusion times.[65]
  • Therapeutic Sequencing: An important clinical question is whether these agents can be used sequentially. Current evidence suggests that the benefit of using one anti-CD38 antibody after a patient has become refractory to the other is limited. Retrospective studies have shown modest response rates when isatuximab is used after daratumumab failure, indicating a degree of class-wide resistance.[67] This finding has significant implications for how to manage patients who relapse on a daratumumab-containing regimen.

B. The Role of Daratumumab in the Modern Therapeutic Armamentarium

  • Establishment as Frontline Standard of Care: The most significant evolution in daratumumab's role has been its migration to the frontline setting. The compelling data from the PERSEUS, CEPHEUS, and ADVANCE trials have firmly established quadruplet regimens, such as D-VRd and D-KRd, as the new standard of care for most newly diagnosed patients, regardless of transplant eligibility.[47] The primary goal of these intensive induction regimens is to achieve the deepest possible response, ideally sustained MRD negativity, from the very beginning of treatment.
  • Moving into Earlier Disease Stages: The therapeutic reach of daratumumab is poised to expand even further into the pre-malignant phase of the disease. The CHMP's positive opinion for its use in high-risk smoldering multiple myeloma (SMM) signals a paradigm shift toward disease interception.[44] The ongoing DETER-SMM trial (NCT03937635) is formally investigating this strategy, aiming to delay or prevent progression to active multiple myeloma in high-risk individuals.[69]

C. Future Perspectives

The success of daratumumab has paved the way for new research questions and therapeutic strategies.

  • Daratumumab Retreatment: With daratumumab now a standard component of frontline therapy, a critical unmet need is how to treat patients who relapse after receiving it. The concept of "daratumumab retreatment" is an area of active investigation. Retrospective analyses suggest that re-challenging with a daratumumab-based regimen, particularly if combined with a novel partner agent and after a sufficient treatment-free interval, can elicit meaningful clinical responses.[57] Prospective trials are needed to define the optimal timing and combination partners for this strategy.
  • Novel Combinations with Next-Generation Immunotherapies: The future of myeloma therapy lies in combining effective agents. Daratumumab is being explored as an ideal backbone partner for a new wave of highly potent immunotherapies, with the goal of achieving synergistic anti-tumor activity.[71] Active clinical trials are investigating its use in combination with:
  • Bispecific T-cell Engagers: Antibodies like teclistamab (targeting BCMA) and talquetamab (targeting GPRC5D) are being combined with daratumumab to engage multiple immune pathways simultaneously.[69]
  • CAR-T Cell Therapy: Daratumumab-based induction regimens are being used to debulk tumors and deepen responses prior to consolidation with CAR-T cell therapy (e.g., ciltacabtagene autoleucel), potentially improving the efficacy and durability of the cellular therapy.[69]
  • Other Novel Agents: Combinations with other targeted agents, such as the BCL-2 inhibitor venetoclax in specific patient subsets, are also being explored.[72]
  • MRD-Guided Therapy: Perhaps the most transformative future application will be the use of MRD status to guide treatment duration. The ability of daratumumab to induce deep, sustained MRD negativity raises the question of whether continuous, indefinite maintenance therapy is necessary for all patients. Future clinical trials will focus on using MRD as a biomarker to de-escalate or even discontinue therapy in patients with a durable molecular remission, thereby personalizing treatment, minimizing long-term toxicity, and improving quality of life.[40]

The very success of daratumumab has created its own next-generation challenge: defining the optimal treatment for the "post-daratumumab" relapse. Its widespread adoption in the frontline setting means that the vast majority of patients who relapse in the future will have been exposed to an anti-CD38 antibody. This fundamentally reshapes the landscape of relapsed/refractory disease, as the highly effective daratumumab-based combinations used in the past may be less effective in this new, pre-exposed population. This reality is the primary driver behind the intense research and development of novel therapies targeting different antigens, such as BCMA and GPRC5D, through platforms like bispecific antibodies and CAR-T cells. These are not just additional drugs; they are the necessary evolution of the field to address the treatment gap created by daratumumab's own transformative impact. The future of myeloma therapy is now inextricably linked to developing effective strategies for patients who progress on a daratumumab-containing regimen.

VIII. Conclusion and Expert Synthesis

Daratumumab has unequivocally revolutionized the management of multiple myeloma and has emerged as a vital new therapy for AL amyloidosis. Its journey from a promising agent for heavily pretreated, refractory patients to a foundational component of frontline, curative-intent strategies has been remarkably rapid and is supported by a wealth of robust evidence from a comprehensive clinical trial program.

The drug's multi-faceted mechanism of action—combining potent, Fc-mediated immune effector functions like CDC, ADCC, and ADCP with direct apoptosis and immunomodulatory effects—provides a powerful and multifaceted attack on CD38-expressing malignant cells. The clinical translation of this mechanism has been profound. Across numerous Phase 3 trials, the addition of daratumumab to standard-of-care backbones has consistently and significantly improved key clinical outcomes, including overall response rates, progression-free survival, and, most importantly, the depth of response.

The ability of daratumumab-based quadruplet regimens to induce unprecedented rates of deep and durable minimal residual disease (MRD) negativity has fundamentally shifted the therapeutic goals in newly diagnosed multiple myeloma. Achieving MRD negativity is no longer a niche research endpoint but is now a central clinical objective, recognized as the strongest prognostic indicator of long-term remission. This has paved the way for the next frontier in myeloma care: MRD-guided therapy, where treatment intensity and duration may one day be tailored to an individual's molecular response. Furthermore, the development of the subcutaneous formulation, Darzalex Faspro, represents a major practical advance, offering patients a more convenient and less burdensome administration route with an improved safety profile regarding infusion reactions, without compromising efficacy.

In conclusion, daratumumab has earned its place as a cornerstone of modern myeloma therapy. It has not only provided a highly effective treatment option that has extended and improved the lives of countless patients but has also raised the bar for therapeutic expectations. In doing so, it has reshaped the natural history of the disease and defined the next great challenge for the field: developing effective strategies for patients in the post-daratumumab era. Its story is a testament to the power of targeted immunotherapy and a paradigm of successful drug development that will continue to influence the field for years to come.

Works cited

  1. Daratumumab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 11, 2025, https://go.drugbank.com/drugs/DB09331
  2. Daratumumab - Wikipedia, accessed July 11, 2025, https://en.wikipedia.org/wiki/Daratumumab
  3. Daratumumab - LiverTox - NCBI Bookshelf, accessed July 11, 2025, https://www.ncbi.nlm.nih.gov/books/NBK548759/
  4. Daratumumab Monograph for Professionals - Drugs.com, accessed July 11, 2025, https://www.drugs.com/monograph/daratumumab.html
  5. Janssen Announces DARZALEX® (daratumumab) U.S. FDA Approval for Newly Diagnosed Patients with Multiple Myeloma who are Transplant Ineligible, accessed July 11, 2025, https://www.jnj.com/media-center/press-releases/janssen-announces-darzalex-daratumumab-us-fda-approval-for-newly-diagnosed-patients-with-multiple-myeloma-who-are-transplant-ineligible
  6. Darzalex (Daratumumab) | International Myeloma Foundation, accessed July 11, 2025, https://www.myeloma.org/darzalex-daratumumab
  7. Darzalex Faspro (daratumumab and hyaluronidase-fihj) FDA ..., accessed July 11, 2025, https://www.drugs.com/history/darzalex-faspro.html
  8. Official Consumer Website | DARZALEX® & DARZALEX FASPRO® (daratumumab and hyaluronidase-fihj), accessed July 11, 2025, https://www.darzalex.com/
  9. www.myeloma.org, accessed July 11, 2025, https://www.myeloma.org/darzalex-daratumumab#:~:text=Darzalex%C2%AE%20(daratumumab)%20is%20the,the%20surface%20of%20myeloma%20cells.
  10. Daratumumab Completed Phase 1 Trials for Multiple Myeloma (MM) Treatment - DrugBank, accessed July 11, 2025, https://go.drugbank.com/drugs/DB09331/clinical_trials?conditions=DBCOND0040908&phase=1&purpose=treatment&status=completed
  11. Daratumumab: a first-in-class CD38 monoclonal antibody for the treatment of multiple myeloma - PMC, accessed July 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4929758/
  12. DARZALEX | Kersten Compliance, accessed July 11, 2025, https://kerstencompliance.com/resources/darzalex
  13. KEGG DRUG: Daratumumab, accessed July 11, 2025, https://www.genome.jp/dbget-bin/www_bget?dr:D10777
  14. Daratumumab - TargetMol, accessed July 11, 2025, https://www.targetmol.com/compound/daratumumab
  15. Clinical Pharmacokinetics and Pharmacodynamics of Daratumumab - PMC, accessed July 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10760401/
  16. DARATUMUMAB - gsrs, accessed July 11, 2025, https://gsrs.ncats.nih.gov/ginas/app/beta/substances/4Z63YK6E0E
  17. Role of daratumumab in the frontline management of multiple myeloma: a narrative review, accessed July 11, 2025, https://www.tandfonline.com/doi/full/10.1080/17474086.2023.2246651
  18. Darzalex, INN-daratumumab, accessed July 11, 2025, https://ec.europa.eu/health/documents/community-register/2022/20220106154122/anx_154122_en.pdf
  19. annex i summary of product characteristics - Darzalex, INN-daratumumab, accessed July 11, 2025, https://ec.europa.eu/health/documents/community-register/2016/20160714135164/anx_135164_en.pdf
  20. Monoclonal Antibody Manufacturing Processes | Avantor, accessed July 11, 2025, https://www.avantorsciences.com/us/en/support/knowledge-center/monoclonal-antibody-production-process
  21. How are Monoclonal Antibodies Produced - AbbVie Contract Manufacturing, accessed July 11, 2025, https://www.abbviecontractmfg.com/news-and-insights/fun-science-friday/how-monoclonal-antibodies-produced.html
  22. kerstencompliance.com, accessed July 11, 2025, https://kerstencompliance.com/resources/darzalex#:~:text=Daratumumab%20active%20substance%20is%20manufactured,viral%20inactivation%2C%20and%20filtration%20steps.
  23. Process Development and Manufacturing for mAbs - Sigma-Aldrich, accessed July 11, 2025, https://www.sigmaaldrich.com/US/en/integrated-offerings/monoclonal-antibodies/process-development-manufacturing
  24. Daratumumab (Darzalex®) in combination with pomalidomide and dexamethasone for the treatment of multiple myeloma (MM), accessed July 11, 2025, https://eprints.aihta.at/1319/1/Oncology%20Fact%20Sheet%20Nr.55_Update.pdf
  25. DARZALEX® (daratumumab) injection 1 HIGHLIGHTS OF PRESCRIBING INFORMATION These highlights do not include all the information n - Janssen, accessed July 11, 2025, https://www.janssenlabels.com/package-insert/product-monograph/prescribing-information/DARZALEX-pi.pdf
  26. DARZALEX FASPRO® (daratumumab and hyaluronidase ... - Janssen, accessed July 11, 2025, https://www.janssenlabels.com/package-insert/product-monograph/prescribing-information/DARZALEX+Faspro-pi.pdf
  27. FDA approves daratumumab and hyaluronidase-fihj for multiple myeloma, accessed July 11, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-daratumumab-and-hyaluronidase-fihj-multiple-myeloma
  28. DARZALEX + DARZALEX FASPRO - Mechanism of Action - J&J Medical Connect, accessed July 11, 2025, https://www.jnjmedicalconnect.com/products/darzalex/medical-content/darzalex-darzalex-faspro-mechanism-of-action
  29. pmc.ncbi.nlm.nih.gov, accessed July 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4929758/#:~:text=Preclinical%20studies%20have%20shown%20that,(ADCP)%2C%20and%20apoptosis.
  30. Role of Daratumumab in Combination With Standard Therapies in Patients With Relapsed and Refractory Multiple Myeloma - PMC - PubMed Central, accessed July 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8204139/
  31. DARZALEX - Pharmacokinetics - J&J Medical Connect, accessed July 11, 2025, https://www.jnjmedicalconnect.com/products/darzalex/medical-content/darzalex-pharmacokinetics
  32. daratumumab - Cancer Care Ontario, accessed July 11, 2025, https://www.cancercareontario.ca/en/drugformulary/drugs/monograph/44446
  33. An update on the role of daratumumab in the treatment of multiple myeloma - PMC, accessed July 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5167079/
  34. 761036Orig1s000 - accessdata.fda.gov, accessed July 11, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/761036Orig1s000ClinPharmR.pdf
  35. DOSING & ADMINISTRATION FOR DARZALEX® (daratumumab), accessed July 11, 2025, https://www.darzalexhcp.com/pdfs/cp-142382v4-darzalex-maia-cassio-apollo-dosing-admin-guide.pdf
  36. Darzalex (daratumumab) FDA Approval History - Drugs.com, accessed July 11, 2025, https://www.drugs.com/history/darzalex.html
  37. Daratumumab (DARZALEX) - FDA, accessed July 11, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/daratumumab-darzalex
  38. DARZALEX - HemOnc.org, accessed July 11, 2025, http://hemonc.org/w/images/archive/a/ad/20161123020856%21Daratumumab.pdf
  39. FDA grants accelerated approval to Darzalex Faspro for newly diagnosed light chain amyloidosis, accessed July 11, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-darzalex-faspro-newly-diagnosed-light-chain-amyloidosis
  40. Darzalex Faspro, Sarclisa Approvals for Multiple Myeloma - NCI, accessed July 11, 2025, https://www.cancer.gov/news-events/cancer-currents-blog/2025/fda-daratumumab-isatuximab-newly-diagnosed-myeloma
  41. FDA approves daratumumab and hyaluronidase-fihj with bortezomib, lenalidomide, and dexamethasone for multiple myeloma, accessed July 11, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-daratumumab-and-hyaluronidase-fihj-bortezomib-lenalidomide-and-dexamethasone-multiple
  42. EMA Review of Daratumumab (Darzalex) for the Treatment of Adult Patients Newly Diagnosed with Multiple Myeloma, accessed July 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7938407/
  43. Darzalex | European Medicines Agency (EMA), accessed July 11, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/darzalex
  44. Darzalex - opinion on variation to marketing authorisation | European Medicines Agency (EMA), accessed July 11, 2025, https://www.ema.europa.eu/en/medicines/human/variation/darzalex
  45. EMA Recommends Extension of Therapeutic Indications for Daratumumab | ESMO, accessed July 11, 2025, https://www.esmo.org/oncology-news/ema-recommends-extension-of-therapeutic-indications-for-daratumumab2
  46. Darzalex Epar Product Information - en | PDF | Natural Killer Cell | Clinical Medicine - Scribd, accessed July 11, 2025, https://www.scribd.com/document/657743407/darzalex-epar-product-information-en
  47. European Commission approves Johnson & Johnson's subcutaneous DARZALEX® (daratumumab)-based quadruplet regimen for the treatment of patients with newly diagnosed multiple myeloma, regardless of transplant eligibility, accessed July 11, 2025, https://www.jnj.com/media-center/press-releases/european-commission-approves-johnson-johnsons-subcutaneous-darzalex-daratumumab-based-quadruplet-regimen-for-the-treatment-of-patients-with-newly-diagnosed-multiple-myeloma-regardless-of-transplant-eligibility
  48. Daratumumab Combo Earns EU Approval in Newly Diagnosed ..., accessed July 11, 2025, https://www.cancernetwork.com/view/daratumumab-combo-earns-eu-approval-in-newly-diagnosed-multiple-myeloma
  49. Darzalex (daratumumab) Receives Positive Opinion from European CHMP for High-Risk Smoldering Multiple Myeloma Patients, accessed July 11, 2025, https://www.myeloma.org/news-events/multiple-myeloma-news/chmp-positive-opinion-darzalex-daratumumab-hrsmm
  50. Daratumumab Plus Standard Therapy for Multiple Myeloma - NCI, accessed July 11, 2025, https://www.cancer.gov/news-events/cancer-currents-blog/2024/multiple-myeloma-darzalex-plus-vrd
  51. Daratumumab, VELCADE (Bortezomib), Lenalidomide and Dexamethasone Compared to VELCADE, Lenalidomide and Dexamethasone in Subjects With Previously Untreated Multiple Myeloma (Perseus) - ClinicalTrials.gov, accessed July 11, 2025, https://www.clinicaltrials.gov/study/NCT03710603
  52. Clinical Results | DARZALEX® & DARZALEX FASPRO® (daratumumab and hyaluronidase-fihj), accessed July 11, 2025, https://www.darzalex.com/clinical-results/
  53. daratumumab 20mg/mL concentrate for solution for infusion and 1,800mg solution for injection (Darzalex®) - Scottish Medicines Consortium, accessed July 11, 2025, https://scottishmedicines.org.uk/media/6870/daratumumab-darzelex-final-april-2022-for-website.pdf
  54. New analyses reinforce long-term benefit of DARZALEX® (daratumumab) subcutaneous-based quadruplet regimen for patients with newly diagnosed multiple myeloma, accessed July 11, 2025, https://innovativemedicine.jnj.com/emea/newsroom/new-analyses-reinforce-long-term-benefit-of-darzalex-daratumumab-subcutaneous-based-quadruplet-regimen-for-patients-with-newly-diagnosed-multiple-myeloma
  55. Daratumumab Combo Improves Outcomes in Transplant-Ineligible NDMM Subgroups, accessed July 11, 2025, https://www.cancernetwork.com/view/daratumumab-combo-improves-outcomes-in-transplant-ineligible-ndmm-subgroups
  56. Daratumumab for patients with myeloma with early or late relapse after initial therapy: subgroup analysis of CASTOR and POLLUX | Blood Advances - ASH Publications, accessed July 11, 2025, https://ashpublications.org/bloodadvances/article/8/2/388/506492/Daratumumab-for-patients-with-myeloma-with-early
  57. Is There a Role for Daratumumab Retreatment in Patients with ..., accessed July 11, 2025, https://www.mdpi.com/2227-9059/13/1/207
  58. New Standard-of-Care Emerges for Multiple Myeloma - InventUM, accessed July 11, 2025, https://news.med.miami.edu/new-standard-of-care-emerges-for-multiple-myeloma/
  59. Future Directions After AURIGA: Daratumumab and Lenalidomide in Myeloma, accessed July 11, 2025, https://www.targetedonc.com/view/future-directions-after-auriga-daratumumab-and-lenalidomide-in-myeloma
  60. How Is Daratumumab Used to Treat Myeloma? - Patient Power, accessed July 11, 2025, https://www.patientpower.info/video/multiple-myeloma/ask-the-expert/how-is-daratumumab-used-to-treat-myeloma
  61. Clinical Policy: Daratumumab (Darzalex), Daratumumab/Hyaluronidase-fihj (Darzalex Faspro) - Louisiana Department of Health, accessed July 11, 2025, https://ldh.la.gov/assets/medicaid/PharmPC/5.17.22/LA.PHAR.310DaratumumabDarzalex04.22.pdf
  62. Darzalex (daratumumab) vs Sarclisa (isatuximab) | Everyone.org, accessed July 11, 2025, https://everyone.org/explore/compare?id1=43&id2=390
  63. Daratumumab, isatuximab (CD38 drugs) - Science for ME, accessed July 11, 2025, https://www.s4me.info/threads/daratumumab-isatuximab-cd38-drugs.42556/
  64. Isatuximab in the Treatment of Multiple Myeloma: A Review and Comparison With Daratumumab - PMC, accessed July 11, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9340383/
  65. Assessing CD38 Monoclonal Antibody-Based Regimens in NDMM - CancerNetwork, accessed July 11, 2025, https://www.cancernetwork.com/view/assessing-cd38-monoclonal-antibody-based-regimens-in-ndmm
  66. Clinical Perspectives: Selecting Between Daratumumab- and Isatuximab-Containing Quadruplet Induction Regimens - CancerNetwork, accessed July 11, 2025, https://www.cancernetwork.com/view/clinical-perspectives-selecting-between-daratumumab-and-isatuximab-containing-quadruplet-induction-regimens
  67. Daratumumab vs isatuximab in the treatment of multiple myeloma - YouTube, accessed July 11, 2025, https://www.youtube.com/watch?v=w1popgmRJwA
  68. Real-world clinical outcomes in patients with multiple myeloma treated with isatuximab after daratumumab treatment - PubMed, accessed July 11, 2025, https://pubmed.ncbi.nlm.nih.gov/37115297/
  69. Clinical Trials Using Daratumumab - NCI, accessed July 11, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/daratumumab?pn=1
  70. Study Details | Daratumumab Retreatment in Participants With Multiple Myeloma Who Have Been Previously Treated With Daratumumab | ClinicalTrials.gov, accessed July 11, 2025, https://clinicaltrials.gov/study/NCT03871829
  71. The future of daratumumab in multiple myeloma - YouTube, accessed July 11, 2025, https://www.youtube.com/watch?v=a9mnXkzl4oI
  72. A Study of Combination Therapy With Venetoclax, Daratumumab and Dexamethasone (With and Without Bortezomib) in Participants With Relapsed or Refractory Multiple Myeloma | ClinicalTrials.gov, accessed July 11, 2025, https://www.clinicaltrials.gov/study/NCT03314181?term=abt-199%20myeloma&viewType=Table&rank=6

Published at: July 11, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.