Cracking the Code to Successful Stem Cell Mobilization in Multiple Myeloma

i3health 2,366 views 44 slides Oct 08, 2024
Slide 1
Slide 1 of 44
Slide 1
1
Slide 2
2
Slide 3
3
Slide 4
4
Slide 5
5
Slide 6
6
Slide 7
7
Slide 8
8
Slide 9
9
Slide 10
10
Slide 11
11
Slide 12
12
Slide 13
13
Slide 14
14
Slide 15
15
Slide 16
16
Slide 17
17
Slide 18
18
Slide 19
19
Slide 20
20
Slide 21
21
Slide 22
22
Slide 23
23
Slide 24
24
Slide 25
25
Slide 26
26
Slide 27
27
Slide 28
28
Slide 29
29
Slide 30
30
Slide 31
31
Slide 32
32
Slide 33
33
Slide 34
34
Slide 35
35
Slide 36
36
Slide 37
37
Slide 38
38
Slide 39
39
Slide 40
40
Slide 41
41
Slide 42
42
Slide 43
43
Slide 44
44

About This Presentation

i3 Health is pleased to make the speaker slides from this activity available for use as a non-accredited self-study or teaching resource.

This slide deck will explore strategies for successful stem cell mobilization in multiple myeloma, focusing on optimizing peripheral blood stem cell collection f...


Slide Content

Hamza Hashmi, MD Assistant Attending, Myeloma & Cell Therapy Service Memorial Sloan Kettering Cancer Center Assistant Professor of Clinical Medicine Weill Cornell Medical College Cracking the Code to Successful Stem Cell Mobilization in Multiple Myeloma

Disclosures A dvisory board/panel: Janssen S peaker's bureau: Janssen, Karyopharm i3 Health has mitigated all relevant financial relationships

Learning Objectives MM = multiple myeloma: ASCT = autologous hematopoietic stem cell transplantation. Evaluate current and emerging data on stem cell mobilization regimens in patients with MM Identify patients eligible for ASCT and at high risk of mobilization failure Apply interdisciplinary strategies to mitigate poor mobilization and optimize ASCT outcomes

Case Study: Mr. AJ PET = positron emission tomography; CT = computed tomography. 65-year-old African-American male presents with 1-month history of right rib pain, not relieved by over-the-counter analgesics Labs reveal hemoglobin 9.5 g/dL, creatinine 2.1 mg/dL, calcium 12.5 mg/dL, monoclonal M-protein 3.5 g/dL, IgA kappa, serum kappa free light chain 120 mg/L, lambda free light chain 1.1 mg/L, kappa lambda light chain ratio >100 Whole-body PET/CT revealed multiple lytic lesions in the axial and appendicular skeleton with a compression fracture at T8 vertebrae. Bone marrow biopsy revealed 80% kappa-restricted plasma cells

Case Study: Mr. AJ (cont.) RXT = radiotherapy; D-RVd = daratumumab/lenalidomide/bortezomib/dexamethasone; BMBx = bone marrow biopsy; BMT = bone marrow transplantation. Patient receives 30 Gy of palliative RXT to L5 spine and pelvis for pain control followed by 8 cycles of D-RVd induction chemotherapy with 25 mg of lenalidomide BMBx reveals residual 20% clonal plasma cells. Whole-body PET/CT is without evidence of active myeloma bone disease Patient is evaluated by BMT service with plan for f ilgrastim ± plerixafor- based steady-state mobilization

MM Background BM = bone marrow; OS = overall survival; IMiDs = immunomodulatory drugs; PIs = proteasome inhibitors; CD38 = cluster of differentiation 38; mAbs = monoclonal antibodies. SEER, 2024; Siegel et al, 2024; Raab et al, 2009; Kumar et al, 2008; Palumbo et al, 2011; Pasvolsky et al, 2023. MM is characterized by malignant plasma cells that accumulate in the BM, leading to bone destruction, organ damage, and bone marrow failure Median age for diagnosis of MM is 69 years The American Cancer Society estimates 35,780 new MM cases and 12,540 deaths in the United States in 2024 MM is the second most common hematologic malignancy, historically associated with median OS of 24-30 months The development of high-dose chemotherapy and ASCT, IMiDs, PIs, anti-CD38 mAbs, and other novel therapies has greatly expanded therapeutic options for newly diagnosed MM Currently, median OS exceeds 45-82 months, with ASCT playing a pivotal role in the treatment paradigm for MM

Approach to Transplant- Eligible NDMM NDMM = newly diagnosed MM; PS = performance status; Dara = daratumumab; VRD = bortezomib/lenalidomide/dexamethasone; KRd = carfilzomib/lenalidomide/dexamethasone; MSK ABMT SOP = Memorial Sloan Kettering Adult BMT Service Standard Operating Procedure; CR = complete response; HDT = high-dose therapy; KR = carfilzomib/lenalidomide; VD-PACE = bortezomib/dexamethasone/cisplatin/doxorubicin/cyclophosphamide/etoposide; EMD = extramedullary disease; PCL = plasma cell leukemia; auto = autologous; allo = allogeneic. Voorhees et al, 2023; Sonneveld et al, 2023; Gay et al, 2021; Gay et al, 2023; McCarthy et al, 2017; Nooka et al, 2014; Dimopoulos et al, 2018; Usmani et al, 2021; Giralt et al, 2014; Ghandili et al, 2023; Maffini et al, 2019. Consider VD-PACE for bulky EMD or PCL cytoreduction Consider planned auto/allo for ultra–high-risk Stem cell mobilization; adequate stem cell harvest (≥8×10 6 CD34+ cells/kg) as per MSK ABMT SOP HDT-ASCT ASCT-eligible patients Patients with good PS and adequate organ function Dara-VRd x4 cycles Dara- KRd x4 cycles Standard-risk High-risk Consider consolidation with induction regimen for patients who do not achieve CR or better Lenalidomide ± dara maintenance KR maintenance

ASCT in MM Background PFS = progression-free survival; CD34 = cluster of differentiation 34; PBSC = peripheral blood stem cell; HSPC = hematopoietic stem and progenitor cells. Parrondo et al, 2020; Wei & Wei, 2023; Giralt et al, 2014; Giralt et al, 2009. HDT-ASCT has been shown to increase the depth of response, PFS, and OS in eligible MM patients Recommendations for stem cell collection dose: Minimum target: 2 x 10 6 CD34+ cells/kg Recommended: 3-5 x 10 6 CD34+ cells/kg (single) or 8-10 x 10 6 CD34+ cells/kg (2; tandem) CD34+ cell dose plays a role in engraftment and outcomes  accelerated hematological reconstitution (shortened platelet and neutrophil engraftment time), improved OS, and reduced length of hospitalization with higher doses The collection of sufficient autologous PBSCs relies on the successful mobilization of HSPCs from the BM niche into circulation

Pathways in HSPC Mobilization CXCR4 = C-X-C chemokine receptor type 4; SDF-1 = stromal cell-derived factor 1; G-CSF = granulocyte-colony stimulating factor; PTH = parathyroid hormone; HSPG = heparin sulfate proteoglycans; VLA4 = very late antigen-4; VCAM = vascular adhesion cell molecule; S1-P = sphingosine-1-phosphate; EP4 = E-type prostanoid receptor 4; PGE2 = prostaglandin E2; NSAIDs = nonsteroidal anti-inflammatory drugs. Panch et al, 2017. Adhesion molecules retain HSPCs in the BM CXCR4 receptors on HSPCs interact with chemokine SDF-1 A disruption in this interaction causes mobilization of HSPCs Modulated by: G-CSF: SDF-1 downregulation and release of proteases Plerixafor and motixafortide : block CXCR4 binding to SDF-1 Plerixafor OR motixafortide

Factors Associated With Poor Mobilization PB = peripheral blood. Wei & Wei, 2023; Hsu et al, 2023; Giralt et al, 2014; Hulin et al, 2021. Patient- and Disease-Related Older age (>70) Extensive BM involvement Treatment-Related Induction therapy >6 months before stem cell mobilization Prior radiotherapy involving marrow rich sites Multiple lines of chemotherapy (4+) Previous exposure to melphalan, fludarabine, platinum-containing regimens, alkylating agents, lenalidomide, or daratumumab At Time of Mobilization Absolute monocyte count <500/µL Platelet count <150,000/µL Low pre-apheresis PB CD34+ cell count Low Day 1 apheresis yield History of mobilization failure

Current HSPC Mobilization Strategies GM-CSF = granulocyte-macrophage colony stimulating factor. Wei et al, 2023; Crees et al, 2023. Cytokine mobilization alone: G-CSF or GM-CSF Filgrastim ± plerixafor Filgrastim + chemotherapy ± plerixafor Pegfilgrastim + plerixafor Filgrastim + m otixafortide

Mobilization Schedules With Stem Cell Mobilizers SQ = subcutaneous. Shi et al, 2024; Mozobil ® prescribing information, 2023; Aphexda ® prescribing information, 2023. Medication Mobilization Days Primary Collection Days Primary Collection Days Optional Collection Days 1 (Thurs) 2 (Friday) 3 (Sat) 4 (Sun) 5 (Mon) Apheresis session 5 Apheresis session 6 (Tues) Apheresis session 7 (Wed) Apheresis session 8 (Thurs) Apheresis session G-CSF SQ 10 µg/kg in AM (9:00 AM) ALONE OR PLUS: Plerixafor SQ 0.24 mg/kg in PM (9:00 PM) OR 11 hours (or 15-17 hours) prior to initiation of apheresis Motixafortide SQ 1.25 mg/kg in PM (9:00 PM) 10-14 hours prior to initiation of apheresis

Plerixafor SDF-1⍺ = stromal cell-derived factor 1 alpha. Mozobil ® prescribing information, 2023. Plerixafor is an inhibitor of the CXCR4 chemokine receptor and blocks binding of its ligand SDF-1 α , resulting in mobilization of HSPCs from BM into the PB Mechanism of Action In combination with G-CSF to mobilize HSPCs to the PB for collection and subsequent autologous transplantation in patients with MM and non-Hodgkin lymphoma Indication Initiate plerixafor after filgrastim has been administered daily for 4 days  ≤83 kg: 20-mg dose or select dose based on 0.24 mg/kg actual body weight >83 kg: select dose based on 0.24 mg/kg actual body weight, maximum 40 mg/day Administer by SQ injection approximately 11 hours prior to initiation of apheresis (15-17 hours) Renal impairment: if creatinine clearance is ≤50 mL/min, decrease dose by one-third to 0.16 mg/kg, maximum 27 mg/day Dose and Administration

G-CSF + Plerixafor Pivotal Trial: AMD3100-3102 Study 1 According to International Myeloma Working Group (IMWG) Response Criteria. ECOG = Eastern Cooperative Oncology Group; PR = partial response; ITT = intention-to-treat. DiPersio et al, 2009. Trial design (n=302): Prospective, phase 3, double-blind, placebo-controlled Eligibility criteria: 18-78 years of age Confirmed diagnosis of MM ECOG PS 0–1 Adequate organ function undergoing first ASCT in first or second CR or PR 1 n=148 G-CSF (10 µg/kg) SQ Days 1-5 (and 6-8 if needed) Plerixafor (0.24 mg/kg) SQ beginning Day 4 for up to 4 days n=154 G-CSF (10 µg/kg) SQ Days 1-5 (and 6-8 if needed) Placebo 1:1 Primary end point: Proportion of patients collecting ≥6 x 10 6 CD34+ cells/kg within 2 apheresis sessions Secondary end points: Proportion of patients collecting ≥2 x 10 6 CD34+, ≥6 x 10 6 CD34+ cells/kg, within 4 or less apheresis sessions; number of apheresis days required to reach ≥6 x 10 6 CD34+ cells/kg ITT

DiPersio et al, 2009. Mobilization Schedule Medication Mobilization Days Primary Collection Days Optional Collection Days 1 2 3 4 5 Apheresis session 6 Apheresis session 7 Apheresis session 8 Apheresis session G-CSF SQ 10 µg/kg in AM PLUS: Plerixafor SQ 0.24 mg/kg in PM 11 hours prior to apheresis G-CSF + Plerixafor Pivotal Trial: AMD3100-3102 Study (cont.)

DiPersio et al, 2009; Micallef et al, 2018. Proportion With ≥6 x 10 6 CD34+ Cells/kg in ≤2 Apheresis Days Proportion With ≥6 x 10 6 CD34+ Cells/kg in ≤4 Apheresis Days Proportion With ≥2 x 10 6 CD34+ Cells/kg in ≤4 Apheresis Days Median Number of Apheresis Days Median Number of CD34+ Cells/kg G-CSF + plerixafor (n=148) 71.6% 75.7% 95.3% 1.0 10.96 G-CSF + placebo (n=154) 34.4% 51.3% 88.3% 4.0 6.18 P <0.001 P <0.001 P =0.031 P <0.001 P <0.001 The most common adverse events related to plerixafor were gastrointestinal disorders (diarrhea, nausea, vomiting) and injection site reactions (erythema) Long-term follow up with plerixafor showed no difference in OS or PFS with addition of plerixafor at 5 years G-CSF + Plerixafor Pivotal Trial: AMD3100-3102 Study (cont.)

AEs = adverse events. DiPersio et al, 2009. Safety Most Common AEs Related to Study Treatment (≥5% Patients) Plerixafor (n=147) Placebo (n=151) Any related AEs, n(%) 95 (64.6%) 67 (44.4%) Gastrointestinal disorders Diarrhea 27 (18.4%) 8 (5.3%) Nausea 24 (16.3%) 11 (7.3%) Vomiting 8 (5.4%) 4 (2.7%) General disorders and administration site conditions Fatigue 12 (8.2%) 5 (3.3%) Injection site erythema 30 (20.4%) 5 (3.3%) Musculoskeletal and connective tissue disorders Bone pain 14 (9.5%) 12 (7.9%) Nervous system disorders Headache 8 (5.4%) 13 (8.6%) Paresthesia 11 (7.5%) 11 (7.3%) G-CSF + Plerixafor Pivotal Trial: AMD3100-3102 Study (cont.)

Case Study: Mr. AJ (continued) Mr. AJ is slated to collect 5 x 10 6 cells/kg for 2 potential transplants He receives filgrastim 10 µg/kg for 4 consecutive days as well as plerixafor 0.24 mg/kg the night before the first apheresis session Peripheral blood CD34+ count on Days 5, 6, and 7 of apheresis cycle remains low at 5/µL, 8/µL, and 6/µL, with decreasing platelet count on each day. Patient denies any other mobilization-related adverse events Patient collects 1.4 x 10 6 cells/kg after 3 days of apheresis

Lenalidomide and Daratumumab DiPersio et al, 2009; Chhabra et al, 2023. MM plerixafor pivotal trial was conducted prior to the daratumumab/anti-CD38 mAbs and quadruplet induction therapy era A recent post-hoc analysis was done to examine the effect of daratumumab- and lenalidomide-containing quadruplet induction therapy on stem cell mobilization and yields and to evaluate engraftment kinetics to explore potential effects on stem cell recovery in NDMM Data from the MASTER and GRIFFIN trials were analyzed MASTER trial: 4 28-day cycles in the single arm D-KRd: daratumumab plus carfilzomib, lenalidomide, dexamethasone GRIFFIN trial: 4 21-day cycles randomized to: RVd: lenalidomide, bortezomib, dexamethasone D-RVd: daratumumab with RVd Impact of Inclusion in Quadruplet Induction Therapy in NDMM

Chhabra et al, 2023. Impact of Inclusion in Quadruplet Induction Therapy in NDMM Lenalidomide and Daratumumab (cont.)

Lenalidomide and Daratumumab (cont.) DiPersio et al, 2009; Chhabra et al, 2023. Among patients who underwent mobilization: MASTER, D-KRd: 98% (114 of 116)  ASCT, median CD34+ cell dose of 3.2 x 10 6 CD34+ cells/kg GRIFFIN, D-RVd: 99% (94 of 95)  ASCT, median CD34+ cell dose of 4.2 x 10 6 CD34+ cells/kg GRIFFIN, RVd: 98% (78 of 80)  ASCT, median CD34+ cell dose of 4.8 x 10 6 CD34+ cells/kg Patients who collected with G-CSF alone in the first mobilization cycle: 100% (4 of 4) in D-KRd, 89 % (17 of 19) in D-RVd, and 96% (27 of 28) in RVd Impact of Inclusion in Quadruplet Induction Therapy in NDMM

Lenalidomide and Daratumumab (cont.) DiPersio et al, 2009; Chhabra et al, 2023. Patients who did not meet the center-specific minimally required CD34+ cell yield in the first mobilization attempt: MASTER, D- KRd : 7% (8 of 116) 5 patients in upfront plerixafor and 3 patients in rescue plerixafor GRIFFIN: D- RVd 2% (2 of 95) GRIFFIN: RVd 6% (5 of 80) 4 cycles of daratumumab- and lenalidomide-based quadruplet induction therapy had a minimal impact on stem cell mobilization and allowed predictable stem cell harvesting and engraftment in all patients who underwent ASCT Impact of Inclusion in Quadruplet Induction Therapy in NDMM

Plerixafor Impact at Different Peripheral CD34+ Thresholds Shah et al, 2020. Single-center, retrospective analysis included patients with MM and other plasma cell dyscrasias who underwent mobilization with G-CSF followed by autologous stem cell collection between January 2012 and December 2016 Primary end points Evaluation of changes in mean plerixafor utilization and apheresis days and assessment of the impact on overall mobilization costs Secondary end points Description of the impact of lenalidomide use on mobilization and evaluation of the rate of mobilization failure

NS = not specified. Shah et al, 2020. The combined cost of plerixafor and apheresis procedures at a threshold of 40 cells/µL is close to that at a threshold of 15 cells/µL, while saving 26 apheresis days per 100 patients Peripheral Blood CD34+ Threshold to Initiate Plerixafor Mean Plerixafor Doses Per Mobilization Mean Apheresis Days Per Mobilization Calculated Cost Per Mobilization (Apheresis and Plerixafor) < 15 cells/µL n=66 1.32 2.15 $37,506 <20 cells/µL n=130 1.65 2.17 $40,164 <40 cells/µL n=148 1.74 1.89 $37,182 P =0.026 P =0.011 P =NS Peripheral CD34+ Threshold for Plerixafor Administration

Impact Of Lenalidomide Use on Mobilization 1 P values from Kruskal-Wallis test. 2 P value = 0.058: none and ≤6 cycles; <0.001: none and >6 cycles; 0.001: ≤6 and >6 cycles. 3 P value = 0.274: none and ≤6 cycles ; 0.011: none and >6 cycles; 0.04: ≤6 and >6 cycles. 4 P value = 0.013: none and ≤6 cycles; 0.004: none and >6 cycles; 0.197: ≤6 and >6 cycles. Shah et al, 2020. Patients who received more than 6 cycles of lenalidomide demonstrated impaired mobilization and required more apheresis sessions ( P <0.011) and greater plerixafor use ( P <0.001) to achieve target stem cell yields Characteristics Lenalidomide Use None <6 cycles >6 cycles P value 1 Number of patients 74 115 41 Plerixafor doses, mean 1.18 1.46 2.10 <0.001 2 Apheresis days, mean 1.80 1.94 2.34 0.014 3 Day 4 PB CD34+ (median, /µL) 17.40 10.74 9.38 0.003 4 Day 5 PB CD34+ (median, /µL) 58.70 55.13 55.08 0.201 Collection yield (mean, x 10 6 /kg) 8.43 8.74 7.97 0.0136

Impact Of Lenalidomide Use on Mobilization (cont.) Cowan et al, 2021. Single-center, retrospective analysis of patients with MM who attempted stem cell mobilization and collection between January 2012 and July 2015 Prior lenalidomide exposure did not have a statistically significant impact on the absolute number of CD34+ cells collected or on the duration of collection based on a multivariate linear regression analysis for association between receipt of >6 cycles of lenalidomide Linear multivariate analysis showed trend toward decreasing collection yield with increasing cycles of lenalidomide ( P =0.205) Recommendation: refer to autologous mobilization prior to 6 cycles of lenalidomide-based therapy

Stem Cell Mobilization Algorithm Slide courtesy of Hamza Hashmi, MD.   Pegfilgrastim can be given at 6- or 12-mg dose on Day 3 Plerixafor dosing may vary based on weight (<83 kg) and renal function (creatinine clearance <50 mL/min). Review with PharmD Preemptive Steady State Mobilization Pegfilgrastim 6 mg SQ Day 3 (9:00 AM) If using G-CSF instead, use 10 µg/kg starting on Day 4 (not Day 3) Plerixafor 24 mg SQ Day 1 (9:00 PM) PB CD34+ reaches peaks 10-18 (average ~14) hours after plerixafor Tips for Poor Mobilizers If given 6 mg pegfilgrastim , consider daily G-CSF 10 ug/kg at 7 am starting Day 2 Review plerixafor dose with PharmD Revisit collection goal with MD if goal not met For failed collection, remobilize 2-4 weeks later, consider chemomobilization , discuss with group 1 ASCT goal = 2.5 x 10 6 /kg 2 ASCT goal = 5.0 x 10 6 /kg PB CD34+ cell count on Day 1 (7:00 AM) PB CD34+ cell count <5 PB CD34+ cell count 5-10 (for 1 ASCT) or 10-20 (for 2 ASCT) or prior day collection yield <1 x 10 6 /kg PB CD34+ cell count >10 (for 1 ASCT) or >20 (for 2 ASCT) or prior day collection yield ≥1 x 10 6 /kg (1 ASCT) or ≥2 x 10 6 /kg (2 ASCT) Hold apheresis, notify MD, give plerixafor + G-CSF Proceed with apheresis, notify MD, give plerixafor plus G-CSF Proceed with apheresis, give plerixafor Repeat on all days of collection

Motixafortide CXCL12 = C-X-C motif chemokine ligand 12. Aphexda ® prescribing information, 2023. Stem cell mobilizing agent that selectively inhibits CXCR4 binding to SDF-1α/CXCL12, resulting in mobilization of HSPCs from the BM into the PB Mechanism of Action Combination with G-CSF to mobilize HSPCs to the PB for collection and subsequent autologous transplantation in patients with MM Indication Initiate motixafortide after filgrastim has been administered daily for 4 days  Recommended dosage is 1.25 mg/kg by SQ injection 10-14 hours prior to initiation of apheresis A second dose of motixafortide can be administered 10-14 hours before a third apheresis, if necessary Dose and Administration

GENESIS Trial Crees et al, 2023. Trial design (n=122): Prospective, phase 3, double-blind, placebo-controlled, multicenter study Eligibility criteria: 18-78 years of age Confirmed diagnosis of MM ECOG PS 0–1 Adequate organ function undergoing first ASCT in first or second CR or PR n=80 G-CSF (10 µg/kg) SQ Days 1-5 (and 6-8 if needed) Motixafortide (1.25 mg/kg) SQ on Day 4 (and 6 if needed) n=42 G-CSF (10 µg/kg) SQ Days 1-5 (and 6-8 if needed) Placebo 2:1 ITT Objective: assessing the superiority of motixafortide + G-CSF over placebo + G-CSF to mobilize HSPCs for ASCT in MM

GENESIS Trial (cont.) Crees et al, 2023; Aphexda ® prescribing information, 2023. Mobilization Days Medication Mobilization Days Primary Collection Days Optional Collection Days 1 2 3 4 5 Apheresis session 6 Apheresis session 7 Apheresis session 8 Apheresis session G-CSF SQ 10 µg/kg in AM PLUS: Motixafortide SQ 1.25 mg/kg in PM 10-14 hours prior to initiation of apheresis

GENESIS Trial (cont.) CI = confidence interval. Crees et al, 2023. The median number of CD34+ HSPCs mobilized in 1 apheresis day with motixafortide + G-CSF was 10.8 x 10 6 vs 2.25 x 10 6 cells kg –1 with placebo + G-CSF Results: Local Laboratory Assessment Motixafortide + G-CSF (n=80) Placebo + G-CSF (n=42) Odds Ratio (95% CI) P Value Percentage of subjects collecting > 6 x 10 6 CD34+ cells/kg in ≤2 apheresis sessions 92.5% 26.2% 53.3 (14.1, 201.3) <0.0001 Percentage of subjects collecting > 6 x 10 6 CD34+ cells/kg in 1 apheresis session 88.8% 9.5% 118 (25.4, 549.4) <0.0001 Percentage of subjects collecting > 2 x 10 6 CD34+ cells/kg in 1 apheresis sessions 96.3% 64.3% 18.90 (4.5, 80.0) <0.0001

GENESIS Trial (cont.) OR = odds ratio. Crees et al, 2023. Results: Local Laboratory Assessment No patients in the treatment arm had a 4 th day of apheresis

GENESIS Trial (cont.) Aphexda ® prescribing information, 2023. Results: Central Laboratory Assessment Motixafortide + G-CSF (n=80) Placebo + G-CSF (n=42) P value Percentage of subjects collecting > 6 x 10 6 CD34+ cells/kg in ≤2 apheresis sessions 67.5% 9.5% <0.0001 Percentage of subjects collecting > 6 x 10 6 CD34+ cells/kg in 1 apheresis session 63.8% 2.4% <0.0001 Percentage of subjects collecting > 2 x 10 6 CD34+ cells/kg in 1 apheresis sessions 87.5% 38.1% <0.0001

GENESIS Trial (cont.) Crees et al, 2023. Time to neutrophil and platelet engraftment, graft durability following transplantation, OS, and PFS were comparable across treatment groups Following administration of either motixafortide or placebo but before apheresis, the median number of PB CD34+ HSPCs in the motixafortide + G-CSF cohort was 116 vs 19 cells μl –1 in the placebo + G‑CSF cohort ( P <0.001) Results

GENESIS Trial (cont.) TEAEs = treatment-emergent adverse events. Crees et al, 2023. TEAEs (Frequency >10%) Motixafortide + G-CSF Placebo + G-CSF Any grade Grade 3 Any grade Grade 3 Total, % (n) 93.8% (75 of 80) 27.5% (22 of 80) 83.3% (35 of 42) 4.8% (2 of 42) Local injection site reactions, % (n) Pain 50% (40 of 80) 6.3% (5 of 80) 4.8% (2 of 42) Erythema 27.5% (22 of 80) Pruritis 21.3% (17 of 80) Systemic injection reactions, % (n) Flushing 32.5% (26 of 80) 7.5% (6 of 80) Pruritis 33.8% (27 of 80) 11.3% (9 of 80) Urticaria 12.5% (10 of 80) 1.3% ( 1 of 80) Erythema 12.5% (10 of 80) Bone pain 17.5% (14 of 80) 31.0% (13 of 42) Back pain 17.5% (14 of 80) 14.3% (6 of 42) Nausea 13.8% (11 of 80) 11.9% (5 of 42) Hypokalemia 13.8% (11 of 80) 11.9% (5 of 42) Catheter site pain 11.3% (9 of 80) 14.3% (6 of 42) Safety and Toxicity

Comparison Slide courtesy of Hamza Hashmi , MD. Plerixafor Motixafortide Pre-medications required No Yes Post-injection monitoring No Yes, 1 hour Resource utilization Self-administration Site administration Adverse events Gastrointestinal disorders (diarrhea, nausea, vomiting) and injection site reactions (erythema) Injection site reactions and systemic injection reactions Renal dose adjustment Yes No Brand vs generic Brand and generic Brand only Cost $ (Generic) $$$ Number of injections Up to 4 injections (Days 4-7) Up to 2 injections (Days 4 and 6) Insurance/payor coverage Coverage ? Coverage

Case Study: Mr. AJ (cont.) IV = intravenous; VGPR = very good partial response. With a failed mobilization attempt, Mr. AJ was advised chemomobilization 3 weeks later Patient receives cyclophosphamide 2 g/m 2 on Day -10, followed by daily filgrastim for 10 days Course was complicated by febrile neutropenia requiring hospitalization and need for IV antibiotics PB CD34+ on Day 1 of apheresis was 25/µL, and patient collected 3.5 x 10 cells/kg on Day 1 and 2.5 x 10 6 on Day 2 of apheresis Patient underwent high-dose chemotherapy followed by stem cell infusion 2 weeks later, achieving VGPR on Day +100 post-BMT

Chemomobilization WBC = white blood cell. Wei & Wei, 2023; Johnsrud et al, 2021; Lazzarino et al, 2001; Barlogie et al, 2007; Whitmill , 2016. Option for PBSC mobilization, especially in patients with active disease as it offers both mobilizing effect and possible antitumor activity Takes a longer time (11-13 days) and requires greater resource utilization Hematologic toxicity is the most common complication reported with chemomobilization, and infection has been observed as the most common non-hematologic toxicity Chemomobilization was associated with more complications (30.0% vs 7.4%; P <0.01), including neutropenic fever, emergency department visits, hospitalizations, and increased cost compared with G-CSF–only mobilization (± plerixafor)

Chemomobilization (cont.) Awan et al, 2012. No standard PB CD34+ count prior to apheresis Measure PB CD34+ cell count daily when the patient’s WBC count recovers to ≥4,000/ μL or from Day +12 (after chemotherapy) onwards (whichever occurs first) 1 2 3-7 10 11 12 13 14 15 Apheresis Apheresis Apheresis Apheresis Chemotherapy G-CSF 10 µg/kg >24 hrs after chemo G-CSF G-CSF G-CSF G-CSF + plerixafor G-CSF + plerixafor G-CSF + plerixafor G-CSF + plerixafor

Current HSPCs Mobilization Strategies Giralt et al, 2014; Giralt et al, 2009; Crees et al, 2023. Cytokine mobilization alone G-CSF ± plerixafor G-CSF + chemotherapy ± p lerixafor G-CSF + motixafortide Place in therapy? First-line mobilization strategies Remobilization options

Key Takeaways Plan for collection after 4-6 cycles of quadruplet-based induction chemotherapy Choice of preemptive vs just-in-time use of plerixafor remains institutional workflow–dependent and should aim for optimal resource utilization For patients with mobilization failure or high risk of failure (risk factors) or need for disease control, chemomobilization should be considered The place of motixafortide for mobilization remains unclear at this time; may be considered for patients with prior failure to mobilize

References Aphexda ™ ( motixafortide ) prescribing information (2023). BioLineRx . Available at: https:// www.aphexda.com /files/prescribing- information.pdf Awan F, Kochuparambil ST, Dermer D, et al (2012). Plerixafor salvage is safe and effective in hard-to-mobilize patients undergoing chemotherapy and filgrastim-based peripheral blood progenitor cell mobilization. J Oncol, 2012:931071. DOI:10.1155/2012/931071 Barlogie B, Anaissie E, van Rhee F, et al (2007). Incorporating bortezomib into upfront treatment for multiple myeloma: early results of total therapy 3. Br J Haematol , 138(2):176-85. DOI:10.1111/j.1365-2141.2007.06639.x Chhabra S, Callander N, Watts NL, et al (2023). Stem cell mobilization yields with daratumumab- and lenalidomide-containing quadruplet induction therapy in newly diagnosed multiple myeloma: findings from the MASTER and GRIFFIN trials. Transplant Cell Ther , 29(3):174.e1-174.e10. DOI:10.1016/j.jtct.2022.11.029 Cowan AJ, Stevenson PA, Green DJ,  et al (2021). Prolonged lenalidomide therapy does not impact autologous peripheral blood stem cell mobilization and collection in multiple myeloma patients: a single-center retrospective analysis. Transplant Cell Ther , 27(8):661.e1-661.e6. DOI:10.1016/j.jtct.2021.04.010 Crees ZD, Rettig MP, Jayasinghe RG, et al (2023). Motixafortide and G-CSF to mobilize hematopoietic stem cells for autologous transplantation in multiple myeloma: a randomized phase 3 trial. Nat Med , 29(4):869-879. DOI:10.1038/s41591-023-02273-z Dimopoulos MA, Dytfeld D, Grosicki S, et al (2018). Elotuzumab plus pomalidomide and dexamethasone for multiple myeloma. N Engl J Med , 379(19):1811-1822. DOI:10.1056/NEJMoa1805762 DiPersio JF, Stadtmauer EA, Nademanee A, et al (2009). Plerixafor and G-CSF versus placebo and G-CSF to mobilize hematopoietic stem cells for autologous stem cell transplantation in patients with multiple myeloma. Blood , 113(23):5720-6. DOI:10.1182/blood-2008-08-174946 Gay F, Musto P, Rota- Scalabrini D, et al (2021). Carfilzomib with cyclophosphamide and dexamethasone or lenalidomide and dexamethasone plus autologous transplantation or carfilzomib plus lenalidomide and dexamethasone, followed by maintenance with carfilzomib plus lenalidomide or lenalidomide alone for patients with newly diagnosed multiple myeloma (FORTE): a randomised , open-label, phase 2 trial. Lancet Oncol , 22(12):1705-1720. DOI:10.1016/S1470-2045(21)00535-0 Gay F, Roeloffzen W, Dimopoulos M, et al (2023). Results of the phase III randomized iskia trial: isatuximab -carfilzomib-lenalidomide-dexamethasone vs carfilzomib-lenalidomide-dexamethasone as pre-transplant induction and post-transplant consolidation in newly diagnosed multiple myeloma patients. Presented at: ASH Annual Meeting. LBA-4. Ghandili S, Alihodzic D, Wiessner C, et al (2023). VTd -PACE and VTd -PACE-like regimens are effective salvage therapies in difficult-to-treat relapsed/refractory multiple myeloma: a single-center experience. Ann Hematol , 102(1):117-124. DOI:10.1007/s00277-022-05027-y

References (cont.) Giralt S, Stadtmauer EA, Harousseau JL, et al (2009). International myeloma working group (IMWG) consensus statement and guidelines regarding the current status of stem cell collection and high-dose therapy for multiple myeloma and the role of plerixafor (AMD 3100). Leukemia , 23(10):1904-12. DOI:10.1038/leu.2009.127 Giralt S, Costa L, Schriber J, et al (2014). Optimizing autologous stem cell mobilization strategies to improve patient outcomes: consensus guidelines and recommendations. Biol Blood Marrow Transplant , 20(3):295-308. DOI:10.1016/j.bbmt.2013.10.013 Hulin C, Offner F, Moreau P, et al (2021). Stem cell yield and transplantation in transplant-eligible newly diagnosed multiple myeloma patients receiving daratumumab + bortezomib/thalidomide/dexamethasone in the phase 3 CASSIOPEIA study. Haematologica , 106(8):2257-2260. DOI:10.3324/haematol.2020.261842 Hsu TL, Tsai CK, Liu CY, et al (2023). Risk factors and outcomes of stem cell mobilization failure in multiple myeloma patients. Transfusion Medicine and Hemotherapy, 50(1):39-50. DOI:10.1159/000525565 Johnsrud A, Ladha A, Muffly L, et al (2021). Stem cell mobilization in multiple myeloma: comparing safety and efficacy of cyclophosphamide +/- plerixafor versus granulocyte colony-stimulating factor +/- plerixafor in the lenalidomide era. Transplant Cell Ther , 27(7):590.e1-590.e8. DOI:10.1016/j.jtct.2021.04.016 Kumar SK, Rajkumar SV, Dispenzieri A, et al (2008). Improved survival in multiple myeloma and the impact of novel therapies. Blood , 111(5):2516-20. DOI:10.1182/blood-2007-10-116129 Lazzarino M, Corso A, Barbarano L, et al (2001). DCEP (dexamethasone, cyclophosphamide, etoposide, and cisplatin) is an effective regimen for peripheral blood stem cell collection in multiple myeloma. Bone Marrow Transplant , 28(9):835-9. DOI:10.1038/sj.bmt.1703240 Maffini E, Storer BE, Sandmaier BM, et al (2019). Long-term follow up of tandem autologous-allogeneic hematopoietic cell transplantation for multiple myeloma. Haematologica , 104(2):380-391. DOI:10.3324/haematol.2018.200253 McCarthy PL, Holstein SA, Petrucci MT, et al (2017). Lenalidomide maintenance after autologous stem-cell transplantation in newly diagnosed multiple myeloma: A Meta-Analysis. J Clin Oncol , 35(29):3279-3289. DOI:10.1200/JCO.2017.72.6679 Micallef IN, Stiff PJ, Nademanee AP, et al (2018). Plerixafor plus granulocyte colony-stimulating factor for patients with non-Hodgkin lymphoma and multiple myeloma: long-term follow-up report. Biol Blood Marrow Transplant , 24(6):1187-1195. DOI:10.1016/j.bbmt.2018.01.039 Mozobil ® (plerixafor) prescribing information (2024). Sanofi. Available at: https:// products.sanofi.us / Mozobil / mozobil.html Nooka AK, Kaufman JL, Muppidi S, et al (2014). Consolidation and maintenance therapy with lenalidomide, bortezomib and dexamethasone (RVD) in high-risk myeloma patients. Leukemia , 28(3):690-3. DOI:10.1038/leu.2013.335

References (cont.) Palumbo A, Anderson K (2011). Multiple myeloma. N Engl J Med , 364(11):1046-60. DOI:10.1056/NEJMra1011442 Panch SR, Szymanski J, Savani BN, et al (2017). Sources of hematopoietic stem and progenitor cells and methods to optimize yields for clinical cell therapy. Biol Blood Marrow Transplan t, 23(8):1241-1249. DOI:10.1016/j.bbmt.2017.05.003 Parrondo RD, Ailawadhi S, Sher T, et al (2020). Autologous stem-cell transplantation for multiple myeloma in the era of novel therapies. JCO Oncol Pract , 16(2):56-66. DOI:10.1200/JOP.19.00335 Pasvolsky O, Ghanem S, Milton DR, et al (2024). Outcomes of autologous stem cell transplantation in patients with ultra-high-risk multiple myeloma. Transplant Cell Ther , 29(12):757-762. DOI:10.1016/j.jtct.2023.08.031 Raab MS, Podar K, Breitkreutz I, et al (2009). Multiple myeloma. Lancet , 374(9686):324-39. DOI:10.1016/S0140-6736(09)60221-X SEER (2024). Cancer stat facts: myeloma. Available at: https:// seer.cancer.gov / statfacts /html/ mulmy.html Shah EE, Young RP, Wong SW, et al (2020). Impact of plerixafor use at different peripheral blood CD34+ thresholds on autologous stem cell collection in patients with multiple myeloma. Biol Blood Marrow Transplant , 26(5):876-883. DOI:10.1016/j.bbmt.2019.11.024 Shi PA, Miller LK, Isola LM (2014). Prospective study of mobilization kinetics up to 18 hours after late-afternoon dosing of plerixafor. Transfusion , 54(5):1263-8. DOI:10.1111/trf.12459 Siegel RL, Giaquinto AN, Jemal A (2024). Cancer statistics, 2024. CA Cancer J Clin , 74(1):12-49. DOI:10.3322/caac.21820 Sonneveld P, Dimopoulos MA, Boccadoro M, et al (2024). Lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med , 390(4):301-313. DOI:10.1056/NEJMoa2312054 Usmani SZ, Hoering A, Ailawadhi S, et al (2021). Bortezomib, lenalidomide, and dexamethasone with or without elotuzumab in patients with untreated, high-risk multiple myeloma (SWOG-1211): primary analysis of a randomised , phase 2 trial. Lancet Haematol , 8(1):e45-e54. DOI:10.1016/S2352-3026(20)30354-9 Voorhees PM, Sborov DW, Laubach J, et al (2023). Addition of daratumumab to lenalidomide, bortezomib, and dexamethasone for transplantation-eligible patients with newly diagnosed multiple myeloma (GRIFFIN): final analysis of an open-label, randomised , phase 2 trial. Lancet Haematol , 10(10):e825-e837. DOI:10.1016/S2352-3026(23)00217-X Wei X & Wei Y (2023). Stem cell mobilization in multiple myeloma: challenges, strategies, and current developments. An Hematol , 102(5):995-1009. DOI:10.1007/s00277-023-05170-0 Whitmill R, Lewis D, Sutton D, et al (2015). A retrospective single center comparison of cyclophosphamide plus G-CSF versus G-CSF alone for peripheral blood stem cell (PBSC) mobilisation following first line therapy in patients with multiple myeloma. Blood, 126(23): 1898. DOI:10.1182/blood.V126.23.1898.1898