Pharmacotherapeutic group: Antineoplastic agents, monoclonal antibodies.
ATC code: L01FC01.
Pharmacology: Pharmacodynamics: DARZALEX FASPRO subcutaneous formulation contains recombinant human hyaluronidase (rHuPH20). rHuPH20 works locally and transiently to degrade hyaluronan ((HA), a naturally occurring glycosaminoglycan found throughout the body) in the extracellular matrix of the subcutaneous space by cleaving the linkage between the two sugars (N-acetylglucosamine and glucuronic acid) which comprise HA. rHuPH20 has a half-life in skin of less than 30 minutes. Hyaluronan levels in subcutaneous tissue return to normal within 24 to 48 hours because of the rapid biosynthesis of hyaluronan.
Mechanism of action: Daratumumab is an IgG1κ human monoclonal antibody (mAb) that binds to the CD38 protein expressed on the surface of cells in a variety of hematological malignancies, including clonal plasma cells in multiple myeloma and AL amyloidosis, as well as other cell types and tissues. CD38 protein has multiple functions such as receptor mediated adhesion, signaling and enzymatic activity.
Daratumumab has been shown to potently inhibit the
in vivo growth of CD38-expressing tumor cells. Based on
in vitro studies, daratumumab may utilize multiple effector functions, resulting in immune mediated tumor cell death. These studies suggest that daratumumab can induce tumor cell lysis through complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP) in malignancies expressing CD38. A subset of myeloid derived suppressor cells (CD38+MDSCs), regulatory T cells (CD38+T
regs) and B cells (CD38+B
regs) are decreased by daratumumab. T cells (CD3+, CD4+, and CD8+) are also known to express CD38 depending on the stage of development and the level of activation. Significant increases in CD4+ and CD8+ T cell absolute counts, and percentages of lymphocytes, were observed with DARZALEX FASPRO treatment in peripheral whole blood and bone marrow. T-cell receptor DNA sequencing verified that T-cell clonality was increased with DARZALEX FASPRO treatment, indicating immune modulatory effects that may contribute to clinical response.
Daratumumab induced apoptosis
in vitro after Fc mediated cross-linking. In addition, daratumumab modulated CD38 enzymatic activity, inhibiting the cyclase enzyme activity and stimulating the hydrolase activity. The significance of these
in vitro effects in a clinical setting, and the implications on tumor growth, are not well understood.
Pharmacodynamic effects: Natural killer (NK) cell and T-cell count: NK cells are known to express high levels of CD38 and are susceptible to daratumumab mediated cell lysis. Decreases in absolute counts and percentages of total NK cells (CD16+CD56+) and activated (CD16+CD56
dim) NK cells in peripheral whole blood and bone marrow were observed with DARZALEX FASPRO treatment. However, baseline levels of NK cells did not show an association with clinical response.
Immunogenicity: In multiple myeloma and AL amyloidosis patients treated with DARZALEX FASPRO subcutaneous formulation in monotherapy and combination clinical trials, less than 1% of patients developed treatment-emergent anti-daratumumab antibodies.
In multiple myeloma and AL amyloidosis patients, the incidence of treatment-emergent non-neutralizing anti-rHuPH20 antibodies was 7.1% (58/812) in monotherapy and combination DARZALEX FASPRO clinical trials. The anti-rHuPH20 antibodies did not appear to impact daratumumab exposures. The clinical relevance of the development of anti-daratumumab or anti-rHuPH20 antibodies after treatment with DARZALEX FASPRO subcutaneous formulation is not known.
Immunogenicity data are highly dependent on the sensitivity and specificity of the test methods used. Additionally, the observed incidence of a positive result in a test method may be influenced by several factors, including sample handling, timing of sample collection, drug interference, concomitant medication and the underlying disease. Therefore, comparison of the incidence of antibodies to daratumumab with the incidence of antibodies to other products may be misleading.
Cardiac electrophysiology: Daratumumab as a large protein has a low likelihood of direct ion channel interactions. The effect of daratumumab on the QTc interval was evaluated in an open-label study for 83 patients (Study GEN501) with relapsed and refractory multiple myeloma following daratumumab infusions (4 to 24 mg/kg). Linear mixed PK-PD analyses indicated no large increase in mean QTcF interval (i.e. greater than 20 ms) at daratumumab C
max.
Clinical studies: Clinical experience with DARZALEX FASPRO subcutaneous formulation: Monotherapy-relapsed/refractory multiple myeloma: MMY3012, an open-label, randomized, Phase 3 non-inferiority study, compared efficacy and safety of treatment with DARZALEX FASPRO subcutaneous formulation (1800 mg) vs. intravenous (16 mg/kg) daratumumab in patients with relapsed or refractory multiple myeloma who had received at least 3 prior lines of therapy including a proteasome inhibitor and an immunomodulatory agent or who were double-refractory to a proteasome inhibitor and an immunomodulatory agent. Treatment continued until unacceptable toxicity or disease progression.
A total of 522 patients were randomized: 263 to the DARZALEX FASPRO SC formulation arm and 259 to the IV daratumumab arm. The baseline demographic and disease characteristics were similar between the two treatment groups. The median patient age was 67 years (range: 33-92 years), 55% were male and 78% were Caucasian. The median patient weight was 73 kg (range: 29-138 kg). Patients had received a median of 4 prior lines of therapy. A total of 51% of patients had prior autologous stem cell transplant (ASCT), 100% of patients were previously treated with both PI(s) and IMiD(s) and most patients were refractory to a prior systemic therapy, including both PI and IMiD (49%).
The study was designed to demonstrate non-inferiority of treatment with DARZALEX FASPRO SC formulation versus IV daratumumab based on co-primary endpoints of overall response rate (ORR) by the IMWG response criteria and maximum C
trough at pre-dose Cycle 3 Day 1 (see Pharmacokinetics as follows). The ORR, defined as the proportion of patients who achieve partial response (PR) or better, was 41.1% (95% CI: 35.1%, 47.3%) in the DARZALEX FASPRO SC formulation arm and 37.1% (95% CI: 31.2%, 43.3%) in the IV daratumumab arm.
This study met its primary objectives to show that DARZALEX FASPRO SC formulation is non-inferior to IV daratumumab in terms of ORR and maximum trough concentration. The results are provided in Table 1. (See Table 1.)
Click on icon to see table/diagram/image
After a median follow-up of 29.3 months, the median overall survival (OS) was 28.2 months (95% CI: 22.8, NE) in the DARZALEX FASPRO formulation arm and was 25.6 months (95% CI: 22.1, NE) in the IV daratumumab arm.
Safety and tolerability results, including in lower weight patients, were consistent with the known safety profile for DARZALEX FASPRO SC formulation and IV daratumumab.
Results from the modified-CTSQ, a patient reported outcome questionnaire that assesses patient satisfaction with their therapy, demonstrated that patients receiving DARZALEX FASPRO subcutaneous formulation had greater satisfaction with their therapy compared with patients receiving IV daratumumab.
Combination treatments in multiple myeloma: MMY2040 was an open-label trial evaluating the efficacy and safety of DARZALEX FASPRO SC formulation 1800 mg: D-VMP arm: In combination with bortezomib, melphalan, and prednisone (D-VMP) in patients with newly diagnosed multiple myeloma (MM) who are ineligible for transplant. Bortezomib was administered by subcutaneous (SC) injection at a dose of 1.3 mg/m
2 body surface area twice weekly at Weeks 1, 2, 4 and 5 for the first 6-week cycle (Cycle 1; 8 doses), followed by once weekly administrations at Weeks 1, 2, 4 and 5 for eight more 6-week cycles (Cycles 2-9; 4 doses per cycle). Melphalan at 9 mg/m
2, and prednisone at 60 mg/m
2 were orally administered on Days 1 to 4 of the nine 6-week cycles (Cycles 1-9). DARZALEX FASPRO SC formulation was continued until disease progression or unacceptable toxicity. The median duration of follow-up for patients was 6.9 months.
The median age was 75 years and approximately 51% were ≥75 years of age. The sex of the patients was evenly distributed. Most patients were white (69%). 33% had ISS Stage I, 45% had ISS Stage II, and 22% had ISS Stage III disease at screening.
D-Rd arm: In combination with lenalidomide and dexamethasone (D-Rd) in patients with relapsed or refractory MM. Lenalidomide (25 mg once daily orally on Days 1-21 of repeated 28-day [4-week] cycles) was given with low dose dexamethasone 40 mg/week (or a reduced dose of 20 mg/week for patients >75 years or BMI <18.5). DARZALEX FASPRO subcutaneous formulation was continued until disease progression or unacceptable toxicity. The median duration of follow-up for patients was 7.1 months.
The median age was 69 years. The majority of patients were male (69%). Most patients were white (69%). 42% had ISS Stage I, 30% had ISS Stage II, and 28% had ISS Stage III disease at screening. Patients had received a median of 1 prior line of therapy, 52% of patients received prior autologous stem cell transplantation (ASCT). The majority of patients (95%) received prior PI, 59% received a prior Immunomodulatory Agent including 22% who received prior lenalidomide. 54% of patients received both a prior PI and Immunomodulatory Agents.
D-Kd arm: In combination with carfilzomib and dexamethasone (D-Kd) for patients in first relapse or refractory MM after initial treatment with a lenalidomide-containing regimen. Carfilzomib was administered by IV infusion at a dose of 20 mg/m
2 on Cycle 1 Day 1. If a dose of 20 mg/m
2 was tolerated, carfilzomib was administered at a dose of 70 mg/m
2 as a 30-minute IV infusion, on Cycle 1 Day 8 and Day 15, and then Day 1, 8 and 15 of each cycle. This was given with low-dose dexamethasone 40 mg per week (or a reduced dose of 20 mg per week for patients ≥75 years or BMI <18.5). DARZALEX FASPRO SC formulation was continued until disease progression or unacceptable toxicity. The median duration of follow-up for patients was 9.2 months.
The median age was 61 years and 52% were male. Most patients were white (73%). 68% had ISS Stage I, 18% had ISS Stage II, and 14% had ISS Stage III disease at screening. A total of 79% of patients had received prior ASCT; 91% of patients received prior PI. All patients received 1 prior line of therapy with exposure to lenalidomide and 62% of patients were refractory to lenalidomide.
A total of 198 patients (D-VMP: 67; D-Rd: 65; D-Kd: 66) were enrolled. Efficacy results were determined by computer algorithm using IMWG response criteria. Primary endpoints ORR for D-VMP, D-Rd and D-Kd and VGPR were met (see Table 2). The minimal residual disease (MRD) negativity rate for patients in the D-Kd arm was 24%, based on all treated populations and a threshold of 10
-5. (See Table 2.)
Click on icon to see table/diagram/image
Combination treatment with pomalidomide and dexamethasone in patients with multiple myeloma: Study MMY3013 was an open-label, randomized, active-controlled Phase 3 trial that compared treatment with DARZALEX FASPRO SC formulation (1800 mg) in combination with pomalidomide and low-dose dexamethasone (D-Pd) to treatment with pomalidomide and low-dose dexamethasone (Pd) in patients with multiple myeloma who had received at least one prior therapy with lenalidomide and a protease inhibitor (PI). Pomalidomide (4 mg once daily orally on Days 1-21 of repeated 28-day [4-week] cycles) was given with low dose oral or intravenous dexamethasone 40 mg/week (or a reduced dose of 20 mg/week for patients >75 years). On DARZALEX FASPRO SC formulation administration days, 20 mg of the dexamethasone dose was given as a pre-administration medication and the remainder given the day after the administration. For patients on a reduced dexamethasone dose, the entire 20 mg dose was given as a DARZALEX FASPRO SC formulation pre-administration medication. Dose adjustments for pomalidomide and dexamethasone were applied according to manufacturer's prescribing information. Treatment was continued in both arms until disease progression or unacceptable toxicity.
A total of 304 patients were randomized: 151 to the D-Pd arm and 153 to the Pd arm. The baseline demographic and disease characteristics were similar between the two treatment groups. The median patient age was 67 years (range 35 to 90 years), 18% were ≥75 years, 53% were male, and 89% Caucasian. Patients had received a median of 2 prior lines of therapy, with 11% of patients having received one prior line of therapy. All patients received a prior treatment with a proteasome inhibitor (PI) and lenalidomide, and 56% of patients received prior stem cell transplantation (ASCT). The majority of patients were refractory to lenalidomide (80%), a PI (48%), or both an immunomodulator and a PI (42%). Efficacy was evaluated by PFS based on IMWG criteria.
With a median follow-up of 16.9 months, the primary analysis of PFS in Study MMY3013 demonstrated a statistically significant improvement in the D-Pd arm as compared to the Pd arm; the median PFS was 12.4 months in the D-Pd arm and 6.9 months in the Pd arm (HR [95% CI]: 0.63 [0.47, 0.85]; p-value = 0.0018), representing a 37% reduction in the risk of disease progression or death for patients treated with D-Pd versus Pd. Median OS was not reached for either treatment group. (See Figure 1.)
Click on icon to see table/diagram/image
Additional efficacy results from Study MMY3013 are presented in Table 3 as follows. (See Table 3.)
Click on icon to see table/diagram/image
In responders, the median time to response was 1 month (range: 0.9 to 9.1 months) in the D-Pd group and 1.9 months (range: 0.9 to 17.3 months) in the Pd group. The median duration of response had not been reached in the D-Pd group (range: 1 to 34.9+ months) and was 15.9 months (range: 1+ to 24.8 months) in the Pd group.
Patients treated with D-Pd reported a reduction in pain severity as measured with the EORTC QLQ-C30 and maintained baseline health-related quality of life, symptoms, and functioning for the other EORTC QLQ-C30 and EORTC QLQ-MY20 subscales. These benefits were not observed in patients treated with Pd.
Combination treatment with bortezomib, cyclophosphamide and dexamethasone in patients with AL amyloidosis: Study AMY3001, an open-label, randomized, active-controlled Phase 3 study, compared treatment with DARZALEX FASPRO subcutaneous formulation (1800 mg) in combination with bortezomib, cyclophosphamide and dexamethasone (D-VCd) to treatment with bortezomib, cyclophosphamide and dexamethasone (VCd) alone in patients with newly diagnosed AL amyloidosis. Randomization was stratified by AL amyloidosis Cardiac Staging System, countries that typically offer autologous stem cell transplant (ASCT) for patients with AL amyloidosis, and renal function.
All patients enrolled in study AMY3001 had newly diagnosed AL amyloidosis with at least one affected organ, measurable hematologic disease, cardiac stage I-IIIA (based on European Modification of Mayo 2004 cardiac stage), and NYHA class I-IIIA. Patients with NYHA class IIIB and IV were excluded.
Bortezomib (SC; 1.3 mg/m
2 body surface area), cyclophosphamide (oral or IV; 300 mg/m
2 body surface area; max dose 500 mg), and dexamethasone (oral or IV; 40 mg or a reduced dose of 20 mg for patients >70 years or body mass index [BMI] <18.5 or those who have hypervolemia, poorly controlled diabetes mellitus or prior intolerance to steroid therapy) were administered weekly on Days 1, 8, 15, and 22 of repeated 28-day [4-week] cycles. On the days of DARZALEX FASPRO dosing, 20 mg of the dexamethasone dose was given as a pre-injection medication and the remainder given the day after DARZALEX FASPRO administration. Bortezomib, cyclophosphamide and dexamethasone were given for six 28-day [4-week] cycles in both treatment arms, while DARZALEX FASPRO treatment was continued until disease progression, start of subsequent therapy, or a maximum of 24 cycles (~2 years) from the first dose of study treatment. Dose adjustments for bortezomib, cyclophosphamide and dexamethasone were applied according to manufacturer's prescribing information.
A total of 388 patients were randomized: 195 to the D-VCd arm and 193 to the VCd arm. The baseline demographic and disease characteristics were similar between the two treatment groups. The majority (79%) of patients had lambda free light chain disease. The median patient age was 64 years (range: 34 to 87); 47% were ≥65 years; 58% were male; 76% Caucasian, 17% Asian, and 3% African American; 23% had AL amyloidosis Clinical Cardiac Stage I, 40% had Stage II, 35% had Stage IIIA, and 2% had Stage IIIB. The median number of organs involved was 2 (range: 1-6) and 66% of patients had 2 or more organs involved. Vital organ involvement was: 71% cardiac, 59% renal and 8% hepatic. The major efficacy outcome measure was hematologic complete response (HemCR) rate as determined by the Independent Review Committee assessment based on International Concensus Criteria. Study AMY3001 demonstrated an improvement in HemCR in the D-VCd arm as compared to the VCd arm. Efficacy results are summarized in Table 4. (See Table 4.)
Click on icon to see table/diagram/image
In responders, the median time to HemCR was 60 days (range: 8 to 299 days) in the D-VCd group and 85 days (range: 14 to 340 days) in the VCd group. The median time to VGPR or better was 17 days (range: 5 to 336 days) in the D-VCd group and 25 days (range: 8 to 171 days) in the VCd group. The median duration of HemCR had not been reached in either arm. (See Figure 2.)
Click on icon to see table/diagram/image
The median follow-up for the study is 11.4 months. The median major organ deterioration progression-free survival (MOD-PFS) was not reached for patients in either arm. The median major organ deterioration event-free survival (MOD-EFS) was not reached for patients receiving D-VCd and was 8.8 months for patients receiving VCd. The hazard ratio for MOD-EFS was 0.39 (95% CI: 0.27, 0.56) and the nominal p-value was <0.0001. Overall survival (OS) data were not mature. A total of 56 deaths were observed [N=27 (13.8%) D-VCd vs. N=29 (15%) VCd group].
Patients treated with D-VCd reported clinically meaningful improvement in fatigue and Global Health Status compared with VCd at week 16 of treatment, assessed using EORTC QLQ-C30 (European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30-items). After 6 cycles of treatment, there were meaningful improvements in patients HRQoL (health-related quality of life) outcomes with continued daratumumab treatment. No adjustments were made for multiplicity.
Clinical experience with daratumumab intravenous formulation: Newly Diagnosed Multiple Myeloma Eligible for ASCT: Combination with bortezomib, thalidomide and dexamethasone in patients eligible for autologous stem cell transplant (ASCT): Study MMY3006, an open-label, randomized, active-controlled Phase 3 study compared induction and consolidation treatment with IV daratumumab 16 mg/kg in combination with bortezomib, thalidomide and dexamethasone (D-VTd) to treatment with bortezomib, thalidomide and dexamethasone (VTd) in patients with newly diagnosed multiple myeloma eligible for ASCT. The consolidation phase of treatment began a minimum of 30 days post-ASCT, when the patient had recovered sufficiently, and engraftment was complete.
Bortezomib was administered by subcutaneous (SC) injection or intravenous (IV) injection at a dose of 1.3 mg/m
2 body surface area twice weekly for two weeks (Days 1, 4, 8, and 11) of repeated 28-day (4-week) induction treatment cycles (Cycles 1-4) and two consolidation cycles (Cycles 5 and 6) following ASCT after Cycle 4. Thalidomide was administered orally at 100 mg daily during the six bortezomib cycles. Dexamethasone (oral or intravenous) was administered at 40 mg on Days 1, 2, 8, 9, 15, 16, 22 and 23 of Cycles 1 and 2, and at 40 mg on Days 1-2 and 20 mg on subsequent dosing days (Days 8, 9, 15, 16) of Cycles 3-4. Dexamethasone 20 mg was administered on Days 1, 2, 8, 9, 15, 16 in Cycles 5 and 6. On the days of IV daratumumab infusion, the dexamethasone dose was administered intravenously as a pre-infusion medication. Dose adjustments for bortezomib, thalidomide and dexamethasone were applied according to manufacturer's prescribing information.
A total of 1085 patients were randomized: 543 to the D-VTd arm and 542 to the VTd arm. The baseline demographic and disease characteristics were similar between the two treatment groups. The median age was 58 (range: 22 to 65 years). The majority were male (59%), 48% had an ECOG performance score of 0, 42% had an ECOG performance score of 1 and 10% had an ECOG performance score of 2. Forty percent had ISS Stage I, 45% had ISS Stage II and 15% had ISS Stage III disease.
Efficacy was evaluated by the stringent Complete Response (sCR) rate at Day 100 post-transplant. (See Table 5.)
Click on icon to see table/diagram/image
With a median follow-up of 18.8 months, the primary analysis of PFS in study MMY3006 demonstrated an improvement in PFS in the D-VTd arm as compared to the VTd arm; the median PFS had not been reached in either arm. Treatment with D-VTd resulted in a reduction in the risk of progression or death by 53% compared to VTd alone (HR=0.47; 95% CI: 0.33, 0.67; p<0.0001). Results of an updated PFS analysis after a median follow-up of 44.5 months continued to show an improvement in PFS for patients in the D-VTd arm compared with the VTd arm. Median PFS was not reached in the D-VTd arm and was 51.5 months in the VTd arm (HR=0.58; 95% CI: 0.47, 0.71; p<0.0001), representing a 42% reduction in the risk of disease progression or death in patients treated with D-VTd.
Newly Diagnosed Multiple Myeloma Ineligible for ASCT: Combination treatment with lenalidomide and dexamethasone in patients ineligible for autologous stem cell transplant: Study MMY3008 an open-label, randomized, active-controlled Phase 3 study, compared treatment with IV daratumumab 16 mg/kg in combination with lenalidomide and low-dose dexamethasone (D-Rd) to treatment with lenalidomide and low-dose dexamethasone (Rd) in patients with newly diagnosed multiple myeloma. Lenalidomide (25 mg once daily orally on Days 1-21 of repeated 28-day [4-week] cycles) was given with low dose oral or intravenous dexamethasone 40 mg/week (or a reduced dose of 20 mg/week for patients >75 years or body mass index [BMI] <18.5). On IV daratumumab infusion days, the dexamethasone dose was given as a pre-infusion medication. Dose adjustments for lenalidomide and dexamethasone were applied according to manufacturer's prescribing information. Treatment was continued in both arms until disease progression or unacceptable toxicity.
A total of 737 patients were randomized: 368 to the D-Rd arm and 369 to the Rd arm. The baseline demographic and disease characteristics were similar between the two treatment groups. The median age was 73 (range: 45-90) years, with 44% of the patients ≥75 years of age. The majority were white (92%), male (52%), 34% had an Eastern Cooperative Oncology Group (ECOG) performance score of 0, 50% had an ECOG performance score of 1 and 17% had an ECOG performance score of ≥2. Twenty-seven percent had International Staging System (ISS) Stage I, 43% had ISS Stage II and 29% had ISS Stage III disease. Efficacy was evaluated by progression free survival (PFS) based on International Myeloma Working Group (IMWG) criteria and overall survival (OS).
With a median follow-up of 28 months, the primary analysis of PFS in study MMY3008 demonstrated an improvement in the D-Rd arm as compared to the Rd arm; the median PFS had not been reached in the D-Rd arm and was 31.9 months in the Rd arm (hazard ratio [HR]=0.56; 95% CI: 0.43, 0.73; p<0.0001), representing 44% reduction in the risk of disease progression or death in patients treated with D-Rd. Results of an updated PFS analysis after a median follow-up of 64 months continued to show an improvement in PFS for patients in the D-Rd arm compared with the Rd arm. Median PFS was 61.9 months in the D-Rd arm and 34.4 months in the Rd arm (HR=0.55; 95% CI: 0.45, 0.67; p<0.0001), representing a 45% reduction in the risk of disease progression or death in patients treated with D-Rd.
After a median follow-up of 56 months, D-Rd has shown an OS advantage over the Rd arm (HR=0.68; 95% CI: 0.53, 0.86; p=0.0013), representing a 32% reduction in the risk of death in patients treated in the D-Rd arm. Median OS was not reached for either arm. The 60-month survival rate was 66% (95% CI: 61, 71) in the D-Rd arm and was 53% (95% CI: 47, 59) in the Rd arm.
In responders, the median time to response was 1.05 months (range: 0.2 to 12.1 months) in the D-Rd group and 1.05 months (range: 0.3 to 15.3 months) in the Rd group. The median duration of response had not been reached in the D-Rd group and was 34.7 months (95% CI: 30.8, not estimable) in the Rd group.
Additional efficacy results from Study MMY3008 are presented in Table 6 as follows.
Combination treatment with bortezomib, melphalan and prednisone (VMP) in patients ineligible for autologous stem cell transplant: Study MMY3007, an open-label, randomized, active-controlled Phase 3 study, compared treatment with IV daratumumab 16 mg/kg in combination with bortezomib, melphalan and prednisone (D-VMP), to treatment with VMP in patients with newly diagnosed multiple myeloma. Bortezomib was administered by subcutaneous (SC) injection at a dose of 1.3 mg/m
2 body surface area twice weekly at Weeks 1, 2, 4 and 5 for the first 6-week cycle (Cycle 1; 8 doses), followed by once weekly administrations at Weeks 1, 2, 4 and 5 for eight more 6-week cycles (Cycles 2-9; 4 doses per cycle). Melphalan at 9 mg/m
2 and prednisone at 60 mg/m
2 were orally administered on Days 1 to 4 of the nine 6-week cycles (Cycles 1-9). IV daratumumab treatment was continued until disease progression or unacceptable toxicity.
A total of 706 patients were randomized: 350 to the D-VMP arm and 356 to the VMP arm. The baseline demographic and disease characteristics were similar between the two treatment groups. The median age was 71 (range: 40-93) years, with 30% of the patients ≥75 years of age. The majority were white (85%), female (54%), 25% had an ECOG performance score of 0, 50% had an ECOG performance score of 1 and 25% had an ECOG performance score of 2. Patients had IgG/IgA/Light chain myeloma in 64%/22%/10% of instances, 19% had ISS Stage I, 42% had ISS Stage II and 38% had ISS Stage III disease. Efficacy was evaluated by PFS based on IMWG criteria and overall survival (OS).
With a median follow-up of 16.5 months, the primary analysis of PFS in study MMY3007 demonstrated an improvement in the D-VMP arm as compared to the VMP arm; the median PFS had not been reached in the D-VMP arm and was 18.1 months in the VMP arm (HR=0.5; 95% CI: 0.38, 0.65; p<0.0001), representing 50% reduction in the risk of disease progression or death in patients treated with D-VMP. Results of an updated PFS analysis after a median follow-up of 40 months continued to show an improvement in PFS for patients in the D-VMP arm compared with the VMP arm. Median PFS was 36.4 months in the D-VMP arm and 19.3 months in the VMP arm (HR=0.42; 95% CI: 0.34, 0.51; p<0.0001), representing a 58% reduction in the risk of disease progression or death in patients treated with D-VMP.
After a median follow-up of 40 months, D-VMP has shown an overall survival (OS) advantage over the VMP arm (HR=0.60; 95% CI: 0.46, 0.80; p=0.0003), representing a 40% reduction in the risk of death in patients treated in the D-VMP arm. Median OS was not reached for either arm.
In responders, the median time to response was 0.79 months (range: 0.4 to 15.5 months) in the D-VMP group and 0.82 months (range: 0.7 to 12.6 months) in the VMP group. The median duration of response had not been reached in the D-VMP group and was 21.3 months (range: 18.4, not estimable) in the VMP group.
Additional efficacy results from Study MMY3007 are presented in Table 6 as follows.
Relapsed/Refractory Multiple Myeloma: Combination treatment with lenalidomide and dexamethasone: Study MMY3003, an open-label, randomized, active-controlled Phase 3 trial, compared treatment with IV daratumumab 16 mg/kg in combination with lenalidomide and low-dose dexamethasone (D-Rd) to treatment with lenalidomide and low-dose dexamethasone (Rd) in patients with multiple myeloma who had received at least one prior therapy. Lenalidomide (25 mg once daily orally on Days 1-21 of repeated 28-day [4-week] cycles) was given with low dose oral or intravenous dexamethasone 40 mg/week (or a reduced dose of 20 mg/week for patients >75 years or BMI <18.5). On IV daratumumab infusion days, 20 mg of the dexamethasone dose was given as a pre-infusion medication and the remainder given the day after the infusion. For patients on a reduced dexamethasone dose, the entire 20 mg dose was given as an IV daratumumab pre-infusion medication. Dose adjustments for lenalidomide and dexamethasone were applied according to manufacturer's prescribing information. Treatment was continued in both arms until disease progression or unacceptable toxicity.
A total of 569 patients were randomized; 286 to the D-Rd arm and 283 to the Rd arm. The baseline demographic and disease characteristics were similar between the IV daratumumab and the control arm. The median patient age was 65 years (range 34 to 89 years), 11% were ≥75 years, 59% were male; 69% Caucasian, 18% Asian, and 3% African American. Patients had received a median of 1 prior line of therapy. Sixty-three percent (63%) of patients had received prior autologous stem cell transplantation (ASCT). The majority of patients (86%) received a prior proteasome inhibitor (PI), 55% of patients had received a prior immunomodulatory agent (IMiD), including 18% of patients who had received prior lenalidomide, and 44% of patients had received both a prior PI and IMiD. At baseline, 27% of patients were refractory to the last line of treatment. Eighteen percent (18%) of patients were refractory to a PI only, and 21% were refractory to bortezomib. Efficacy was evaluated by PFS based on IMWG criteria and overall survival (OS).
With a median follow-up of 13.5 months, the primary analysis of PFS in study MMY3003 demonstrated an improvement in the D-Rd arm as compared to the Rd arm; the median PFS had not been reached in the D-Rd arm and was 18.4 months in the Rd arm (HR=0.37; 95% CI: 0.27, 0.52; p<0.0001) representing 63% reduction in the risk of disease progression or death in patients treated with D-Rd. Results of an updated PFS analysis after a median follow-up of 55 months continued to show an improvement in PFS for patients in the D-Rd arm compared with the Rd arm. Median PFS was 45.0 months in the D-Rd arm and 17.5 months in the Rd arm (HR=0.44; 95% CI: 0.35, 0.54; p<0.0001), representing a 56% reduction in the risk of disease progression or death in patients treated with D-Rd.
After a median follow-up of 80 months, D-Rd has shown an OS advantage over the Rd arm (HR=0.73; 95% CI: 0.58, 0.91; p=0.0044), representing a 27% reduction in the risk of death in patients treated in the D-Rd arm. The median OS was 67.6 months in the D-Rd arm and 51.8 months in the Rd arm. The 78-month survival rate was 47% (95% CI: 41, 52) in the DRd arm and was 35% (95% CI: 30, 41) in the Rd arm.
Additional efficacy results from Study MMY3003 are presented in Table 6 as follows.
Combination treatment with bortezomib and dexamethasone: Study MMY3004, an open-label, randomized, active-controlled Phase 3 trial, compared treatment with IV daratumumab 16 mg/kg in combination with bortezomib and dexamethasone (D-Vd), to treatment with bortezomib and dexamethasone (Vd) in patients with multiple myeloma who had received at least one prior therapy. Bortezomib was administered by SC injection or IV injection at a dose of 1.3 mg/m
2 body surface area twice weekly for two weeks (Days 1, 4, 8, and 11) of repeated 21 day (3-week) treatment cycles, for a total of 8 cycles. Dexamethasone was administered orally at a dose of 20 mg on Days 1, 2, 4, 5, 8, 9, 11, and 12 of the 8 bortezomib cycles (80 mg/week for two out of three weeks of each of the bortezomib cycle) or a reduced dose of 20 mg/week for patients >75 years, BMI <18.5, poorly controlled diabetes mellitus or prior intolerance to steroid therapy. On the days of IV daratumumab infusion, 20 mg of the dexamethasone dose was administered as a pre-infusion medication. For patients on a reduced dexamethasone dose, the entire 20 mg dose was given as an IV daratumumab pre-infusion medication. Bortezomib and dexamethasone were given for 8 three-week cycles in both treatment arms; whereas IV daratumumab was given until treatment progression. However, dexamethasone 20 mg was continued as an IV daratumumab pre-infusion medication in the D-Vd arm. Dose adjustments for bortezomib and dexamethasone were applied according to manufacturer's prescribing information.
A total of 498 patients were randomized; 251 to the D-Vd arm and 247 to the Vd arm. The baseline demographic and disease characteristics were similar between the IV daratumumab and the control arm. The median patient age was 64 years (range 30 to 88 years); 12% were ≥75 years, 57% were male; 87% Caucasian, 5% Asian and 4% African American. Patients had received a median of 2 prior lines of therapy and 61% of patients had received prior autologous stem cell transplantation (ASCT). Sixty-nine percent (69%) of patients had received a prior PI (66% received bortezomib) and 76% of patients received an IMiD (42% received lenalidomide). At baseline, 32% of patients were refractory to the last line of treatment and the proportions of patients refractory to any specific prior therapy were well balanced between the treatment groups. Thirty-three percent (33%) of patients were refractory to an IMiD only, and 28% were refractory to lenalidomide. Efficacy was evaluated by PFS based on IMWG criteria and overall survival (OS).
With a median follow-up of 7.4 months, the primary analysis of PFS in study MMY3004 demonstrated an improvement in the D-Vd arm as compared to the Vd arm; the median PFS had not been reached in the D-Vd arm and was 7.2 months in the Vd arm (HR [95% CI]: 0.39 [0.28, 0.53]; p-value <0.0001), representing a 61% reduction in the risk of disease progression or death for patients treated with D-Vd versus Vd. Results of an updated PFS analysis after a median follow-up of 50 months continued to show an improvement in PFS for patients in the D-Vd arm compared with the Vd arm. Median PFS was 16.7 months in the D-Vd arm and 7.1 months in the Vd arm (HR [95% CI]: 0.31 [0.24, 0.39]; p-value <0.0001), representing a 69% reduction in the risk of disease progression or death in patients treated with D-Vd versus Vd.
After a median follow-up of 73 months, D-Vd has shown an OS advantage over the Vd arm (HR=0.74: 95% CI: 0.59, 0.92; p=0.0075), representing a 26% reduction in the risk of death in patients treated in the D-Vd arm. The median OS was 49.6 months in the D-Vd arm and 38.5 months in the Vd arm. The 72-month survival rate was 39% (95% CI: 33, 45) in the D-Vd arm and was 25% (95% CI: 20, 31) in the Vd arm.
Additional efficacy results from Study MMY3004 are presented in Table 6 as follows. (See Table 6.)
Click on icon to see table/diagram/image
Combination treatment with pomalidomide and dexamethasone: Study MMY1001 was an open-label trial in which 103 patients with multiple myeloma who had received a prior PI and an IMiD, received IV daratumumab 16 mg/kg in combination with pomalidomide and low-dose dexamethasone until disease progression. Pomalidomide (4 mg once daily orally on Days 1-21 of repeated 28-day [4-week] cycles) was given with low dose oral or intravenous dexamethasone at 40 mg/week (reduced dose of 20 mg/week for patients >75 years or BMI <18.5). On IV daratumumab infusion days, 20 mg of the dexamethasone dose was given as a pre-infusion medication and the remainder given the day after the infusion. For patients on a reduced dexamethasone dose, the entire 20 mg dose was given as an IV daratumumab pre-infusion medication.
The median patient age was 64 years (range: 35 to 86 years) with 8% of patients ≥75 years of age. Patients in the study had received a median of 4 prior lines of therapy. Seventy-four percent (74%) of patients had received prior ASCT. Ninety eight percent (98%) of patients received prior bortezomib treatment, and 33% of patients received prior carfilzomib. All patients received prior lenalidomide treatment, with 98% of patients previously treated with the combination of bortezomib and lenalidomide. Eighty nine percent (89%) of patients were refractory to lenalidomide and 71% refractory to bortezomib; 64% of patients were refractory to bortezomib and lenalidomide.
Overall response rate was 59% (95% CI: 49.1%, 68.8%); VGPR or better was achieved in 42% of patients, CR or better was achieved in 14% of patients and stringent CR was achieved in 8% of patients. The Clinical Benefit Rate (ORR+ MR [Minimal response]) was 62% (95% CI: 52.0, 71.5). The median time to response was 1 month (range: 0.9 to 2.8 months). The median duration of response was 13.6 months (95% CI: 10.0, not estimable). After a median duration of follow-up of 9.8 months, the median OS was not reached. The 12-month survival rate was 72%.
Combination treatment with twice-weekly (20/56 mg/m
2) carfilzomib and dexamethasone: Study 20160275, an open-label, randomized, active-controlled Phase 3 trial, compared treatment with DARZALEX 16 mg/kg in combination with carfilzomib and dexamethasone (DKd) to treatment with carfilzomib and dexamethasone (Kd) in patients with multiple myeloma who had received at least one to three prior lines of therapy.
Carfilzomib was administered as IV infusion twice weekly on Days 1, 2, 8, 9, 15, and 16 of repeated 28-day [4-week] treatment cycles. Carfilzomib dose was 20 mg/m
2 on Cycle 1 Days 1 and 2 and 56 mg/m
2 beginning on Cycle 1 Day 8 and thereafter.
Dexamethasone was administered orally or by IV infusion at a dose of 40 mg weekly. Dexamethasone was administered as an IV infusion on carfilzomib and/or DARZALEX IV infusion days. On the days of DARZALEX and/or carfilzomib infusion, 20 mg of the dexamethasone dose was administered via IV as a pre-infusion medication. The remaining 20 mg of dexamethasone was administered via IV on successive day of carfilzomib and/or DARZALEX infusions. For patients >75 years on a reduced dexamethasone dose of 20 mg, the entire 20 mg dose was given as a DARZALEX pre-infusion medication. Dose adjustments for carfilzomib and dexamethasone were applied according to manufacturer's prescribing information. Treatment was continued in both arms until disease progression or unacceptable toxicity.
A total of 466 patients were randomized; 312 to the DKd arm and 154 to the Kd arm. The baseline demographic and disease characteristics were similar between the DARZALEX and the control arm. The median patient age was 64 years (range 29 to 84 years), 9% were ≥75 years, 58% were male; 79% Caucasian, 14% Asian, and 2% African American. Patients had received a median of 2 prior lines of therapy and 58% of patients had received prior autologous stem cell transplantation (ASCT). The majority of patients (92%) received a prior PI and of those 34% were refractory to PI including regimen. Fourty-two percent (42%) of patients had received prior lenalidomide and of those, 33% were refractory to a lenalidomide containing regimen.
Efficacy was evaluated by PFS based on IMWG criteria. Study 20160275 demonstrated an improvement in PFS in the DKd arm as compared to the Kd arm; the median PFS had not been reached in the DKd arm and was 15.8 months in the Kd arm (hazard ratio [HR]=0.630; 95% CI: 0.464, 0.854; p=0.0014), representing 37% reduction in the risk of disease progression or death for patients treated with DKd versus Kd. PFS improvement observed in the ITT population was also observed in lenalidomide refractory patients. (See Figure 3.)
Click on icon to see table/diagram/image
After a median follow-up of 17.1 months, 95 deaths were observed [N=59 (19%) in the DKd group and N=36 (23%) in the Kd group]. Overall survival (OS) data were not mature, however, there was a trend toward longer OS in the DKd arm compared with the Kd arm (HR=0.745; 95% CI: 0.491, 1.131; p=0.0836).
Additional efficacy results from Study 20160275 are presented in the table as follows. (See Table 7.)
Click on icon to see table/diagram/image
In responders, the median time to response was 1 month (range: 1 to 14 months) in the DKd group and 1 month (range: 1 to 10 months) in the Kd group. The median duration of response had not been reached in the DKd group and was 16.6 months (95% CI: 13.9, not estimable) in the Kd group.
Combination treatment with once-weekly (20/70 mg/m
2) carfilzomib and dexamethasone: Study MMY1001 was an open-label trial in which 85 patients with multiple myeloma who had received at least one prior therapy, received 16 mg/kg DARZALEX in combination with carfilzomib and low-dose dexamethasone until disease progression. Carfilzomib was administered as IV infusion once weekly at a dose of 20 mg/m
2 on Cycle 1 Day 1 and escalated to dose of 70 mg/m
2 on Cycle 1 Days 8 and 15, and Days 1, 8, and 15 of subsequent cycles. Dexamethasone was given at 40 mg (subjects ≤75 years) or 20 mg (subjects >75 years) per week. For first DARZALEX split-dose, dexamethasone was administered on Day 1 and Day 2 before the DARZALEX infusions. During other DARZALEX infusion weeks, dexamethasone was administered on infusion days at a dose of 20 mg before the DARZALEX infusion and 20 mg the day after the DARZALEX infusion.
The median patient age was 66 years (range: 38 to 85 years) with 9% of patients ≥75 years of age. Patients in the study had received a median of 2 prior lines of therapy. Seventy three percent (73%) of patients had received prior ASCT. All patients received prior bortezomib, and 95% of patients received prior lenalidomide. Sixty percent (60%) of patients were refractory to lenalidomide and 29% of patients were refractory to both a PI an IMiD.
Efficacy results were based on overall response rate using IMWG criteria (see Table 8).
Click on icon to see table/diagram/image
The median duration of response was 28 months (95% CI: 20.5, not estimable). The median PFS was 26 months (95% CI: 4.8, NE), after a median follow-up of 24 months. Median overall survival was not reached. The 24-month survival rate was 71%.
Monotherapy: The clinical efficacy and safety of DARZALEX monotherapy for the treatment of patients with relapsed and refractory multiple myeloma whose prior therapy included a proteasome inhibitor and an immunomodulatory agent, was demonstrated in two open-label studies.
In Study MMY2002, 106 patients with relapsed and refractory multiple myeloma received 16 mg/kg DARZALEX until disease progression. The median patient age was 63.5 years (range, 31 to 84 years), 49% were male and 79% were Caucasian. Patients had received a median of 5 prior lines of therapy. Eighty percent of patients had received prior autologous stem cell transplantation (ASCT). Prior therapies included bortezomib (99%), lenalidomide (99%), pomalidomide (63%) and carfilzomib (50%). At baseline, 97% of patients were refractory to the last line of treatment, 95% were refractory to both, a PI and IMiD, 77% were refractory to alkylating agents, 63% were refractory to pomalidomide and 48% of patients were refractory to carfilzomib.
Efficacy results based on Independent Review Committee (IRC) assessment are presented in the table as follows. (See Table 9.)
Click on icon to see table/diagram/image
Overall response rate (ORR) in MMY2002 was similar regardless of type of prior anti-myeloma therapy. With a median duration of follow-up of 9 months, median OS was not reached. The 12-month OS rate was 65% (95% CI: 51.2, 75.5).
In Study GEN501, 42 patients with relapsed and refractory multiple myeloma received 16 mg/kg DARZALEX until disease progression. The median patient age was 64 years (range, 44 to 76 years), 64% were male and 76% were Caucasian. Patients in the study had received a median of 4 prior lines of therapy. Seventy-four percent of patients had received prior ASCT. Prior therapies included bortezomib (100%), lenalidomide (95%), pomalidomide (36%) and carfilzomib (19%). At baseline, 76% of patients were refractory to the last line of treatment, 64% were refractory to both a PI and IMiD, 60% were refractory to alkylating agents, 36% were refractory to pomalidomide and 17% were refractory to carfilzomib.
Treatment with daratumumab at 16 mg/kg led to a 36% ORR with 5% CR and 5% VGPR. The median time to response was 1 (range: 0.5 to 3.2) month. The median duration of response was not reached (95% CI: 5.6 months, not estimable). With a median duration of follow-up of 10 months, median OS was not reached. The 12-month OS rate was 77% (95% CI: 58.0, 88.2).
Pharmacokinetics: Daratumumab exposure in a monotherapy study (MMY3012) in patients with multiple myeloma following the recommended 1800 mg administration of DARZALEX FASPRO SC formulation (weekly for 8 weeks, biweekly for 16 weeks, monthly thereafter) as compared to 16 mg/kg IV daratumumab for the same dosing schedule, showed non-inferiority for the co-primary endpoint of maximum C
trough (Cycle 3 Day 1 pre-dose), with mean ± SD of 593±306 μg/mL compared to 522±226 μg/mL for IV daratumumab, with a geometric mean ratio of 107.93% (90% CI: 95.74-121.67).
Daratumumab exhibits both concentration and time-dependent pharmacokinetics with first order absorption and parallel linear and nonlinear (saturable) elimination that is characteristic of target-mediated clearance. Following the recommended dose of 1800 mg DARZALEX FASPRO SC formulation as monotherapy, peak concentrations (C
max) increased 4.8-fold and total exposure (AUC
0-7 days) increased 5.4-fold from first dose to last weekly dose (8th dose). Highest trough concentrations for DARZALEX FASPRO SC formulation are typically observed at the end of the weekly dosing regimens for both monotherapy and combination therapy.
In patients with multiple myeloma, the simulated trough concentrations following 6 weekly doses of 1800 mg DARZALEX FASPRO for combination therapy were similar to 1800 mg DARZALEX FASPRO monotherapy.
In patients with multiple myeloma, daratumumab exposure in a combination study with pomalidomide and dexamethasone (MMY3013) was similar to that in monotherapy, with the maximum C
trough (Cycle 3 Day 1 pre-dose) mean ± SD of 537±277 μg/mL following the recommended 1800 mg administration of DARZALEX FASPRO SC formulation (weekly for 8 weeks, biweekly for 16 weeks, monthly thereafter).
In a combination study, AMY3001, in patients with AL amyloidosis, the maximum C
trough (Cycle 3 Day 1 pre-dose) was similar to that in multiple myeloma with mean ± SD of 597±232 μg/mL following the recommended 1800 mg administration of DARZALEX FASPRO SC formulation (weekly for 8 weeks, biweekly for 16 weeks, monthly thereafter).
Absorption and Distribution: At the recommended dose of 1800 mg in multiple myeloma patients, the absolute bioavailability of DARZALEX FASPRO SC formulation is 69%, with an absorption rate of 0.012 hour
-1, with peak concentrations occurring at 70 to 72 h (T
max). At the recommended dose of 1800 mg in AL amyloidosis patients, the absolute bioavailability was not estimated, the absorption rate constant was 0.77 day
-1 (8.31% CV) and peak concentrations occurred at 3 days.
In multiple myeloma patients, the modeled mean estimate of the volume of distribution for the central compartment (V1) is 5.25 L (36.9% CV) and peripheral compartment (V2) was 3.78 L in daratumumab monotherapy, and the modeled mean estimate of the volume of distribution for V1 is 4.36 L (28% CV) and V2 was 2.80 L when daratumumab was administered in combination with pomalidomide and dexamethasone. In AL amyloidosis patients, the model estimated apparent volume of distribution after SC administration is 10.8 L (3.1% CV). These results suggest that daratumumab is primarily localized to the vascular system with limited extravascular tissue distribution.
Metabolism and Elimination: Daratumumab is cleared by parallel linear and nonlinear saturable target-mediated clearances. In multiple myeloma patients, the population PK model estimated mean clearance value of daratumumab is 4.96 mL/h (58.7% CV) in daratumumab monotherapy and 4.32 mL/h (43.5% CV) when daratumumab was administered in combination with pomalidomide and dexamethasone. In AL amyloidosis patients, the apparent clearance after SC administration is 210 mL/day (4.1% CV).
In multiple myeloma patients, the model-based geometric mean post hoc estimate for half-life associated with linear elimination is 20.4 days (22.4% CV) in daratumumab monotherapy and 19.7 days (15.3% CV) when daratumumab was administered in combination with pomalidomide and dexamethasone. In AL amyloidosis patients, the model-based geometric mean post hoc estimate for half-life associated with linear elimination is 27.5 days (74.0% CV). For the monotherapy and combination regimens, the steady state is achieved at approximately 5 months into every 4 weeks dosage at the recommended dose and schedule (1800 mg; once weekly for 8 weeks, every 2 weeks for 16 weeks, and then every 4 weeks thereafter).
A population PK analysis, using data from DARZALEX FASPRO SC formulation monotherapy and combination therapy in multiple myeloma patients, was conducted with data from 487 patients who received DARZALEX FASPRO SC formulation and 255 patients who received IV daratumumab. The predicted PK exposures are summarized in Table 10. (See Table 10.)
Click on icon to see table/diagram/image
A population PK analysis, using data from DARZALEX FASPRO SC formulation combination therapy in AL amyloidosis patients, was conducted with data from 211 patients. At the recommended dose of 1,800 mg, predicted daratumumab concentrations were slightly higher, but generally within the same range, in comparison with multiple myeloma patients. (See Table 11.)
Click on icon to see table/diagram/image
Special populations: Age and gender: Based on population PK analyses in patients (33-92 years) receiving monotherapy or various combination therapies, age had no statistically significant effect on the PK of daratumumab. No individualization is necessary for patients on the basis of age.
Gender had a statistically significant effect on PK parameters in patients with multiple myeloma but not in patients with AL amyloidosis. Slightly higher exposure in females were observed than males, but the difference in exposure is not considered clinically meaningful. No individualization is necessary for patients on the basis of gender.
Renal impairment: No formal studies of DARZALEX FASPRO SC formulation in patients with renal impairment have been conducted. Population PK analyses were performed based on pre-existing renal function data in patients with multiple myeloma receiving DARZALEX FASPRO monotherapy or various combination therapies in patients with multiple myeloma or AL amyloidosis and no clinically important differences in exposure to daratumumab were observed between patients with renal impairment and those with normal renal function.
Hepatic impairment: No formal studies of DARZALEX FASPRO SC formulation in patients with hepatic impairment have been conducted. Population PK analyses were performed in patients with multiple myeloma receiving DARZALEX FASPRO SC formulation monotherapy or various combination therapies in patients with multiple myeloma or in AL amyloidosis. No clinically important differences in the exposure to daratumumab were observed between patients with normal hepatic function and mild hepatic impairment. There were very few patients with moderate and severe hepatic impairment to make meaningful conclusions for these populations.
Race: Based on the population PK analyses in patients receiving either DARZALEX FASPRO SC formulation monotherapy or various combination therapies, the daratumumab exposure was similar across races.
Body Weight: The flat-dose administration of DARZALEX FASPRO subcutaneous formulation 1800 mg as monotherapy achieved adequate exposure for all body-weight subgroups. In patients with multiple myeloma, the mean cycle 3 day 1 C
trough in the lower body-weight subgroup (≤65 kg) was 60% higher and in the higher body weight (>85 kg) subgroup, 12% lower than the intravenous daratumumab subgroup. In some patients with body weight >120 kg, lower exposure was observed which may result in reduced efficacy. However, this observation is based on limited number of patients.
In patients with AL amyloidosis, no meaningful differences were observed in C
trough across body weight.
Non-clinical Information: Carcinogenicity and Mutagenicity: No animal studies have been performed to establish the carcinogenic potential of daratumumab. Routine genotoxicity and carcinogenicity studies are generally not applicable to biologic pharmaceuticals as large proteins cannot diffuse into cells and cannot interact with DNA or chromosomal material.
Reproductive Toxicology: No animal studies have been performed to evaluate the potential effects of daratumumab on reproduction or development.
No systemic exposure of hyaluronidase was detected in monkeys given 22,000 U/kg subcutaneously (12 times higher than the human dose) and there were no effects on embryo-fetal development in pregnant mice given 330,000 U/kg hyaluronidase subcutaneously daily during organogenesis, which is 45 times higher than the human dose.
There were no effects on pre- and post-natal development through sexual maturity in offspring of mice treated daily from implantation through lactation with 990,000 U/kg hyaluronidase subcutaneously, which is 134 times higher than the human doses.
Fertility: No animal studies have been performed to determine potential effects on fertility in males or females.