Introduction to
Darzalex Overview of Darzalex
Darzalex, the trade name for daratumumab, is a first‐in‐class, humanized IgG1κ monoclonal antibody targeting the
CD38 antigen highly expressed on
multiple myeloma cells. From its inception as an innovative therapeutic modality, Darzalex quickly came to represent a breakthrough in the treatment of multiple myeloma. It offers a novel mechanism distinct from traditional chemotherapies, helping to overcome drug resistance and improve survival outcomes in both relapsed/refractory and newly diagnosed settings. Darzalex has been administered in various combinations with standard agents—and more recently in subcutaneous formulations to enhance patient convenience and reduce the lengthy infusion times typical of intravenous administration. The widespread use of Darzalex in multiple myeloma is evident from its approved indications in more than 100 countries and its inclusion as a backbone therapy in over 300,000 patient treatments worldwide.
Mechanism of Action
At the molecular level, Darzalex exploits several immune-mediated mechanisms to induce
tumor cell death. Binding to the CD38 antigen on the surface of myeloma cells, Darzalex facilitates multiple pathways of action. It triggers antibody-dependent cellular cytotoxicity (ADCC), recruiting natural killer (NK) cells and macrophages to attack the malignant cells. Additionally, it activates complement-dependent cytotoxicity (CDC), contributing to enhanced tumor lysis. Darzalex can also mediate antibody-dependent cellular phagocytosis (ADCP) and induce direct apoptotic signaling via Fc receptor-mediated crosslinking. The ability of daratumumab not only to kill tumor cells directly but also modulate immune effector cells creates a robust and multifaceted therapeutic effect. This dual activity is critically important in the setting of multiple myeloma where malignant plasma cells have developed resistance to conventional therapies, thereby making Darzalex a cornerstone in modern myeloma management.
Regulatory Approval History
Initial Approval Process
The regulatory journey of Darzalex began with its evaluation as a novel anti‑CD38 agent in the
relapsed/refractory multiple myeloma setting. In November 2015, the U.S. Food and Drug Administration (FDA) granted accelerated approval for Darzalex as monotherapy for patients who had received a minimum of three prior lines of therapy, including exposure to a
proteasome inhibitor and an immunomodulatory agent, or whose disease was double refractory to these treatments. This rapid approval was based on promising phase II trial data that demonstrated an overall response rate of approximately 30% in heavily pretreated patients. Beyond its initial accelerated approval, the FDA authorizations underscored vital safety and efficacy profiles supported by early-phase clinical investigations. This approval not only marked Darzalex as the first CD38-directed antibody approved in the United States for multiple myeloma but also set the stage for subsequent label expansions.
In Europe, the regulatory process was similarly marked by an expedited review under the accelerated assessment framework. On May 20, 2016, the European Commission granted a conditional marketing authorization for Daratumumab as monotherapy in adult patients with relapsed and refractory multiple myeloma who had prior exposure to a proteasome inhibitor and an immunomodulatory agent, or in those who exhibited disease progression after the last treatment. The European Medicines Agency (EMA) reviewed the data from trials that paralleled the U.S. experience, and the positive opinion from the Committee for Medicinal Products for Human Use (CHMP) was a critical milestone. The initial approvals were granted with the understanding that additional confirmatory data would be provided to convert the conditional approvals to full approvals. This accelerated pathway in Europe mirrored the need for innovative therapies in a heavily pretreated patient population facing limited treatment options.
Subsequent Approvals and Indications
Following its initial approval, Darzalex’s clinical development and regulatory journey expanded its scope considerably. In subsequent years, accumulating evidence from phase III pivotal trials led to the approval of Darzalex in combination regimens for multiple myeloma, both in the relapsed/refractory and newly diagnosed settings. One significant extension of the therapeutic label was the approval of Darzalex in combination with
lenalidomide and dexamethasone (commonly abbreviated as D-Rd) in newly diagnosed transplant-ineligible patients. Updated analyses from the MAIA study have shown that the D-Rd regimen not only improves progression-free survival (PFS) but also significantly enhances overall survival, minimal residual disease negativity, and the overall response rate.
Further label expansions included approvals for various combination regimens:
• In the U.S., Darzalex in combination with bortezomib and dexamethasone was approved for patients who had received at least one prior line of therapy. Similarly, combinations with carfilzomib and dexamethasone and with pomalidomide and dexamethasone were approved for relapsed or refractory settings.
• In Europe, similar combinations were eventually authorized after confirmatory data were provided. For instance, the combination of Darzalex with bortezomib, thalidomide, and dexamethasone was approved for newly diagnosed patients who were eligible for autologous stem cell transplant, and the label was extended following the results of randomized phase III trials.
The pathway of label extension also includes the transformation of Darzalex’s intravenous (IV) formulation to a subcutaneous (SC) formulation termed Darzalex Faspro. This new formulation significantly reduced administration time—from multi‐hour IV infusions to five‐minute injections—and offered a more favorable safety profile regarding infusion-related reactions. The subcutaneous formulation has been approved for most of the current indications of Darzalex, further increasing patient convenience and improving treatment adherence. This innovation was achieved by co-formulating daratumumab with recombinant human hyaluronidase PH20 (rHuPH20) using Halozyme’s ENHANZE® drug delivery technology.
Additionally, regulatory authorities have expanded the indications of Darzalex to include treatment in earlier lines of therapy and even in non‐multiple myeloma settings. For example, in January 2021, the FDA approved Darzalex Faspro for the treatment of light chain amyloidosis, further broadening its therapeutic applicability. Ongoing applications and supplementary submissions also aim at using Darzalex in combination regimens, such as with pomalidomide and dexamethasone (D-Pd) in second-line and early relapsed settings, demonstrating the continual evolution of its label based on accumulating clinical data.
Clinical Development Pathway
Clinical Trial Phases
The clinical development of Darzalex has been methodically structured across all clinical trial phases, with each phase contributing to an increasingly robust evidence base.
• Phase I studies primarily focused on establishing safety, tolerability, and pharmacokinetics. Early-phase trials demonstrated that daratumumab, even as a single agent, was well tolerated at doses sufficient to induce anti-tumor activity. These studies provided key pharmacodynamic insights—such as its half-life, optimal dosing schedules, and initial evidence of immune-mediated tumor cell depletion.
• Phase II clinical trials were critical in establishing Darzalex’s efficacy in the relapsed/refractory setting. Pivotal phase II trials, such as those leading to the SIRIUS and GEN501 studies, consistently reported overall response rates around 30% as monotherapy in heavily pretreated patients, thereby justifying accelerated approval. This phase provided the groundwork for the understanding of its safety profile and anti-myeloma activity, offering data that formed the basis for regulatory decisions in both the U.S. FDA and EMA.
• Phase III trials were designed to gauge the efficacy of Darzalex in combination with established myeloma regimens and to demonstrate improvements in clinically relevant endpoints such as progression-free survival and overall survival. Landmark studies—including the MAIA trial for transplant-ineligible patients and the GRIFFIN study for transplant-eligible patients—enabled Darzalex to be incorporated earlier in the treatment algorithm. Data from the MAIA study, for instance, with median follow-ups exceeding 64 months, have shown significant improvements in PFS and overall survival compared to standard-of-care regimens. These confirmatory studies supported the conversion of conditional approvals into full indications and paved the way for further label expansions.
Throughout these clinical phases, a wealth of information was gathered on various dose levels, administration schedules, combination partners, and safety endpoints. Studies were designed using both traditional designs—as well as innovative adaptive designs—to optimize dose escalation and monitor for adverse events in real time. The breadth of studies also included comparative assessments of the intravenous and subcutaneous formulations, facilitating the conversion of the administration route to one that improved patient quality of life, reduced infusion-related reactions, and maintained therapeutic efficacy.
Key Study Results
Key clinical trial findings have been pivotal in shaping the clinical development and subsequent approval history of Darzalex. Early phase studies demonstrated that as monotherapy, daratumumab could achieve a promising overall response rate in patients who were heavily pretreated, establishing its clinical potential as a novel therapy for multiple myeloma.
In subsequent combination studies, the addition of Darzalex to established regimens led to marked improvements in key endpoints:
• The MAIA trial evaluated Darzalex in combination with lenalidomide and dexamethasone (D-Rd) in newly diagnosed, transplant-ineligible patients. The updated analyses reported after a median follow-up of 64.5 months showed significant benefits in progression-free survival as well as minimal residual disease (MRD) negativity rates, with overall survival benefits observed after 73.6 months.
• The GRIFFIN study assessed the efficacy of adding Darzalex to a regimen consisting of bortezomib, lenalidomide, and dexamethasone (VRd) in transplant-eligible patients. The trial revealed enhanced stringent complete response rates, deeper responses, and sustained MRD negativity compared to VRd alone, underscoring the potential of the quadruplet regimen as frontline therapy.
• Studies evaluating the subcutaneous formulation (Darzalex Faspro) demonstrated that the SC route was non-inferior to the IV formulation in terms of overall response rate while significantly reducing administration time and the rate of infusion-related reactions. This innovation not only maintained efficacy but also improved the safety profile and patient convenience, as evidenced by data reported from Phase III trials.
The aggregated data from these studies has shown that combining Darzalex with a variety of agents—whether with proteasome inhibitors (e.g., bortezomib, carfilzomib), immunomodulatory drugs (e.g., lenalidomide, pomalidomide), or corticosteroids (e.g., dexamethasone)—produces a significant additive or even synergistic anti-myeloma effect. The convergence of improved efficacy endpoints (such as response rates, PFS, and overall survival) with manageable safety profiles has been crucial in justifying the broad regulatory approvals and in redefining the standard-of-care for multiple myeloma treatment.
Impact and Future Directions
Clinical Impact and Usage
The clinical impact of Darzalex has been profound across the multiple myeloma treatment continuum. Initially developed for a population with limited options in the relapsed/refractory setting, Darzalex has now evolved into a backbone therapy incorporated in both frontline and subsequent lines of treatment. Its introduction has significantly shifted treatment paradigms, with national and international guidelines now recommending daratumumab-based regimens as preferred or highly recommended options.
Clinicians have welcomed Darzalex not only for its robust efficacy when combined with conventional agents but also for its tolerable safety profile. The immunomodulatory effects, combined with direct anti-myeloma activity, have translated into rapid, deep, and durable responses. Furthermore, the conversion of the administration route—from a time-intensive IV infusion to a convenient subcutaneous injection—has minimized treatment burden on patients and healthcare facilities, reducing patient chair time and lowering the risk for infusion-related adverse events.
The widespread adoption of Darzalex has extended beyond therapy to include its use as a bridge to more definitive treatments such as autologous stem cell transplant (ASCT) in transplant-eligible patients. Its efficacy in reducing tumor burden and achieving MRD negativity is particularly promising, with some studies exploring whether such deep responses might eventually translate into a functional cure for a subset of myeloma patients. Consequently, Darzalex has not only improved clinical outcomes and prolonged survival but has also reshaped clinical practice by becoming integral to modern, combination-based therapeutic regimens.
Future Research and Development
Despite the remarkable success of Darzalex, ongoing research endeavors continue to expand its clinical applications and optimize its use. A key focus of future research is the exploration of more refined combination treatment regimens that may further enhance patient outcomes. Investigational studies are ongoing to evaluate Darzalex in novel combinations, such as with emerging immunotherapeutic agents, checkpoint inhibitors, and next-generation proteasome inhibitors, in hopes of achieving even deeper and more durable responses.
Researchers are also examining the role of Darzalex in earlier disease stages. Studies are underway to assess its efficacy in smoldering multiple myeloma, monoclonal gammopathy of undetermined significance (MGUS), and other plasma cell dyscrasias such as MGRS and AL amyloidosis. The approval of Darzalex Faspro for light chain amyloidosis in January 2021 underscores the potential to broaden its clinical use beyond myeloma to other antibody-mediated disorders. These investigations may lead to paradigm shifts where daratumumab not only treats overt malignancy but also possibly delays disease progression in early or precursor states.
Additionally, the development of the SC formulation has sparked interest in further innovations in drug delivery systems. The success of the ENHANZE® technology in reducing administration time has encouraged pharmaceutical companies to explore additional methods to enhance patient adherence and satisfaction. Ongoing studies aim to optimize dosing schedules, reduce adverse events further, and simplify treatment protocols, which will be particularly beneficial in community and home-based care settings.
Furthermore, adaptive clinical trial designs and real-world evidence studies are increasingly being employed to generate more dynamic data on Darzalex’s performance. These novel methodological approaches will allow for more rapid adjustments to dosing regimens and combination strategies based on patient-specific factors and emerging biomarkers. The integration of personalized medicine approaches, including the use of genomic and proteomic biomarkers to predict response, is likely to personalize therapy even further, thereby maximizing benefit while minimizing unnecessary toxicities.
From a regulatory standpoint, ongoing submissions and variations—such as those for label expansions in combination regimens—signal an exciting future where Darzalex’s therapeutic indications continue to broaden. Collaborative efforts between regulatory agencies and pharmaceutical companies also encourage accelerated review timelines under initiatives like Project Orbis and RTOR, ensuring that promising new data quickly inform updated product labels and clinical practice. Such dynamic processes will likely lead to further approvals in earlier lines of therapy and in other hematologic malignancies or related conditions.
In parallel, health economic evaluations and real-world studies continue to demonstrate the cost-effectiveness of Darzalex-based regimens. These analyses are crucial in informing reimbursement decisions and ensuring that patients have broad access to innovative therapies, even as treatments grow more complex and personalized. Given the high burden of disease associated with multiple myeloma, these factors combine to enhance the overall impact of Darzalex on public health.
Conclusion
In summary, the approval history and clinical development pathway of Darzalex illustrate a journey that is as innovative as it is transformative. Beginning with accelerated approvals in the relapsed/refractory multiple myeloma setting in the United States in November 2015 and conditional marketing authorizations in Europe in May 2016, Darzalex rapidly expanded its indications through robust phase II and phase III clinical trials. Early-phase studies demonstrated its safety and promising monotherapy activity, while subsequent combination studies—such as the MAIA and GRIFFIN trials—established its efficacy in frontline and relapsed settings across both transplant-eligible and ineligible population groups.
The continual evolution from intravenous to subcutaneous formulations through the incorporation of advanced drug delivery technologies has further underscored its clinical versatility, reducing treatment time and adverse events, and enhancing patient quality of life. Today, Darzalex is embedded in national treatment guidelines and represents a cornerstone in multiple myeloma management due to its profound impact on response rates, progression-free survival, and overall survival. Its clinical journey has forged a new paradigm in antibody-based immunotherapy, highlighting the benefits of multi-mechanistic action against malignant cells and setting the stage for future innovations.
Looking forward, research efforts are intensively focused on optimizing combination regimens, extending indications to early disease stages and other hematologic malignancies, and harnessing adaptive trial designs and biomarker-driven strategies to personalize therapy further. With ongoing clinical trials exploring novel therapeutic avenues and regulatory submissions for label expansions, Darzalex’s clinical impact is poised to grow even further. Consequently, its development pathway—from early clinical investigations to widespread regulatory acceptance—serves as a testament to the transformational nature of innovative, mechanism-based therapies in modern oncology.
In conclusion, Darzalex has dramatically redefined the treatment of multiple myeloma. It has not only improved survival outcomes and quality of life for a large number of patients but has also paved the way toward a future where combination therapy, early intervention, and personalized treatment strategies can potentially overcome the historically poor prognosis of this challenging malignancy. The evolution of its development, regulatory milestones, and clinical application demonstrates how scientific ingenuity and collaborative efforts between researchers, clinicians, industry, and regulators can converge to deliver breakthrough therapies that fundamentally change clinical practice.