Treatment Options for Triple-class Refractory Multiple Myeloma

Treatment Options for Triple-class Refractory Multiple Myeloma

Journal Pre-proof Treatment Options for Triple-Class Refractory Multiple Myeloma Joseph Mikhael, MD PII: S2152-2650(19)32008-7 DOI: https://doi.org...

1MB Sizes 0 Downloads 34 Views

Journal Pre-proof Treatment Options for Triple-Class Refractory Multiple Myeloma Joseph Mikhael, MD PII:

S2152-2650(19)32008-7

DOI:

https://doi.org/10.1016/j.clml.2019.09.621

Reference:

CLML 1438

To appear in:

Clinical Lymphoma, Myeloma and Leukemia

Received Date: 7 June 2019 Revised Date:

5 August 2019

Accepted Date: 29 September 2019

Please cite this article as: Mikhael J, Treatment Options for Triple-Class Refractory Multiple Myeloma, Clinical Lymphoma, Myeloma and Leukemia (2019), doi: https://doi.org/10.1016/j.clml.2019.09.621. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 The Author(s). Published by Elsevier Inc.

1

Treatment Options for Triple-Class Refractory Multiple

2

Myeloma

3

Joseph Mikhael, MD

4

Applied Cancer Research and Drug Discovery, Translational Genomics Research

5

Institute (TGen), City of Hope Cancer Center, Phoenix, AZ

6

Word count (2000–10,000 words): 4108

7

Address for correspondence: Dr. Joseph Mikhael, Translational Genomics Research

8

Institute (TGen), 445 N. Fifth Street, Phoenix, AZ 85004

9

E-mail contact: [email protected]

10

Mikhael

11

Abstract (178/250)

12

The advent of new, more effective, and less toxic therapies has revolutionized the

13

management of multiple myeloma in the past decade. Despite the availability of new

14

treatments, the majority of patients with multiple myeloma will become refractory to

15

the therapies that currently comprise the hematologic standard of care for the

16

malignancy: proteasome inhibitors, immunomodulatory agents, and monoclonal

17

antibodies. Moreover, in recent years, a new subset of patients refractory to all 3 of

18

these agents has emerged. This population, in which a clear treatment paradigm

19

remains undefined, is characterized by poor survival outcomes. Current approaches

20

to the treatment of triple-class refractory disease are limited, and include

21

conventional chemotherapy, salvage autologous stem cell transplantation, and

22

recycling prior regimens, each of which have generally short-lived efficacy. It is

23

anticipated that additional agents will be available for triple refractory disease in the

24

near future, namely selinexor, chimeric antigen receptor T cell therapy, and next-

25

generation monoclonal antibodies. The development and further refinement of novel

26

treatments for this subset of patients in the coming years should be considered a key

27

clinical and research priority.

28

Key words: Multiple myeloma, Refractory, Autologous stem cell

29

transplantation, Selinexor, Monoclonal antibodies

Page 2

Mikhael

30

Introduction

31

Multiple myeloma (MM), characterized by the expansion of malignant plasma cells in

32

the bone marrow,1 accounts for approximately 1% of all malignancies and 10% of

33

hematologic malignancies.2 In the past decades, the advent of new, more effective,

34

and less toxic therapies has revolutionized the management of MM.3, 4 The ever-

35

expanding treatment landscape parallels substantial improvements in our

36

understanding of the biology of MM, optimized supportive care strategies, and the

37

refinement of combination treatment regimens.2, 3, 5

38

There are currently 3 drug classes available for the treatment of MM that have

39

shifted treatment paradigms and considerably improved outcomes for patients:

40

proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal

41

antibodies.6, 7 Various approaches to treatment using these agents have been

42

developed and these are widely used in the treatment of MM, often in triplet

43

combinations that involve 2 novel agents plus a steroid; a combination treatment

44

strategy has been adopted that uses a number of different therapies with distinct

45

mechanisms of action.8

46

The use of combination treatments has been a cornerstone of the therapeutic

47

management of MM for decades, but the agents included in these regimens have

48

been refined and have evolved considerably in recent years. For example, although

49

melphalan has served as the backbone for several PI and IMiD combinations for

50

more than 30 years, other agents have replaced alkylating agents as first-choice

51

treatment options for MM. However, melphalan remains commonly used as part of

52

the conditioning regimen in autologous stem cell transplant (ASCT)-eligible patients,9

53

and has recently been validated in a large phase III trial of bortezomib, lenalidomide, Page 3

Mikhael

54

and dexamethasone (RVD) with and without ASCT, demonstrating an improved

55

progression-free survival (PFS) in the transplant arm after high-dose melphalan and

56

2 cycles of RVD.10

57 58

The advent of novel therapeutic strategies, and their inclusion in initial therapeutic

59

regimens,11 has resulted in significant improvements in patient outcomes. A real-

60

world evidence study by Fonseca et al12 found that the percentage of patients with

61

MM receiving novel therapy continuously increased from 8.7% in 2000 to 61.3% in

62

2014. Patients with MM diagnosed after 2010 had better survival outcomes.12 Those

63

diagnosed in 2012 were 1.25 times more likely to survive for 2 years than those

64

diagnosed in 2006.12 Over the 14-year study period, patients with MM showed

65

improved OS, with the 2-year survival gap decreasing at a rate of 3% per year

66

between patients with MM and matched controls.12

67

Despite these advances, the majority of patients with MM will become refractory to

68

PIs, IMiDs, and monoclonal antibodies.13 This can leave clinicians unsure of how to

69

proceed, as fewer therapeutic options remain for heavily pretreated patients who

70

develop more aggressive disease, resulting in poorer outcomes for this patient

71

population.14 A multicenter study enrolled 543 patients with triple-class exposed,

72

IMiD- and PI-refractory MM, who had also been treated with an alkylating agent. The

73

median OS was 13 (95% confidence interval [CI] 11–15) months.15 In a 2016

74

retrospective analysis investigating outcomes in a similar patient population, OS was

75

poor despite the availability of newer agents, with a median OS of approximately

76

8 months.14 Furthermore, a 2018 retrospective analysis demonstrated that patients

77

who were penta-refractory to bortezomib, lenalidomide, carfilzomib, pomalidomide, Page 4

Mikhael

78

and daratumumab had a median OS of only 5.6 months.16 The identification of more

79

effective therapeutic interventions for this patient population has therefore emerged

80

as a key priority for MM research.

Page 5

Mikhael

81

Defining a Treatment Paradigm for Triple-Class

82

Refractory MM

83

Although there is no current standard of care for the treatment of patients with

84

relapsed and/or refractory MM (RRMM),17 combination regimens are generally

85

preferred to monotherapy. This is because combination treatments enable multiple

86

pathways to be targeted to induce prolonged durable responses in patients with

87

MM.18

88

However, once patients with MM have become triple-class refractory, there is even

89

less consensus on what can be defined as a preferred approach to therapy. Reusing

90

or recycling prior treatments to which a patient was previously refractory may be

91

considered, in light of synergistic effects of drug combinations, or due to the

92

presence of clonal evolution that may signify renewed drug sensitivity in later

93

disease.19 Although this option may be considered for patients who had previously

94

exhibited a sufficiently durable response (of at least 6 months) to a given regimen,19

95

this would generally yield low response rates with limited PFS in patients with a

96

short-term remission duration or progression following initial treatment, so other

97

strategies are required.20

98

These key alternative approaches include conventional chemotherapy, salvage

99

ASCT, the novel nuclear export inhibitor, selinexor, along with other novel

100

approaches, such as treatment with chimeric antigen receptor (CAR) T cell therapy,

101

and next-generation antibodies and bispecific T cell engager technology.

102

Conventional Chemotherapy

103

Although not used routinely, conventional chemotherapy can elicit a good (albeit Page 6

Mikhael

104

short-lived) response in patients with RRMM.

105

D(T)-PACE (dexamethasone ± thalidomide, cisplatin, doxorubicin,

106

cyclophosphamide, and etoposide) is a chemotherapeutic regimen that has been

107

evaluated in cases of RRMM, with studies reporting response rates of approximately

108

50%. However, because its toxicity is common and PFS is short, D(T)-PACE is

109

generally considered most effective when used as a bridge to other MM treatment

110

interventions.21

111

Similarly, high-dose cyclophosphamide has shown value as a potential bridge to

112

novel therapies in refractory MM. When used in combination with dexamethasone,

113

high-dose cyclophosphamide was shown to be an efficient rescue regimen in

114

double-refractory MM, with an overall response rate (ORR) of 55%, with 3 patients

115

(15%) achieving complete response (CR). Twelve patients (67%) were treated with

116

further therapies after achieving at least stable disease. The median PFS and OS

117

were 6 and 12 months, respectively.22

Page 7

Mikhael

118

Bendamustine, a bifunctional alkylating agent, has demonstrated efficacy at different

119

stages of MM, including patients with more advanced disease. In a retrospective

120

analysis of individuals with heavily pretreated RRMM, 39 patients received a median

121

of 3 cycles of bendamustine, either as monotherapy or concomitantly with

122

corticosteroids. The results found bendamustine to be effective in these patients,

123

with 3% achieving a very good partial response, 33% partial response, 18% minor

124

response, 26% stable disease, and 20% progressive disease; the median event-free

125

survival and OS were 7 and 17 months, respectively 23 In triple-refractory (100%)

126

and penta-refractory (40%, defined as refractory to one monoclonal antibody, two

127

PIs and two IMiDs) patients refractory to CD38 monoclonal antibodies, those who

128

received bendamustine (n = 15) as their next line of therapy had a median PFS and

129

OS of 3.2 and 9.3 months, respectively.24

130

Doxorubicin, cisplatin, and etoposide are 3 further chemotherapeutic agents that

131

have been evaluated in RRMM. Although the depth and duration of responses to

132

these treatments can be poor when used alone, use of these agents in combination

133

can elicit highly active, durable responses, especially when used in synergistic

134

combinations that also reduce toxicity risk.25

135

In combination with more conventional chemotherapy regimens, histone deacetylase

136

(HDAC) inhibitors have shown promise as a treatment option for patients with

137

triple-class refractory MM. HDACs deacetylate the lysine residues of both histones

138

and non-histone proteins, resulting in histone hyperacetylation and alterations in

139

chromatin structure that cause growth cycle arrest and apoptosis in tumor cells.26

140

Preclinical studies investigating HDAC inhibitors and PIs in MM demonstrated a

141

synergistic effect on tumor cells, which leads to the accumulation of polyubiquitinated

Page 8

Mikhael

142

proteins and activation of apoptosis.27 In the phase III PANORAMA1 trial in patients

143

with RRMM who had received 1 to 3 previous lines of therapy, patients treated with

144

the HDAC inhibitor panobinostat plus bortezomib and dexamethasone showed

145

significantly longer median PFS than those receiving placebo plus bortezomib and

146

dexamethasone (11.99 months [95% CI 10.33–12.94] vs. 8.08 months [7.56–9.23];

147

hazard ratio 0.63, 95% CI 0.52–0.76; P < .0001).28 Similar effects were observed in

148

subgroup analysis of patients who had previously received an IMiD, bortezomib plus

149

an IMiD, or at least 2 lines of treatment including bortezomib and an IMiD.29, 30

150

Panobinostat plus bortezomib and dexamethasone had a tolerable safety profile,

151

with the most frequent grade 3–4 adverse events (AEs) being myelosuppression,

152

diarrhea, asthenia or fatigue, peripheral neuropathy, and pneumonia.28-30 Analysis of

153

patients with RRMM who had experienced a failure with daratumumab (n = 354)

154

showed that patients who switched to chemotherapy (P = .0208) or panobinostat (P

155

= .0298) had received a greater number of therapies than those who switched to

156

elotuzumab-based regimens.31

157

Salvage ASCT

158

ASCT was first used in combination with melphalan in patients with MM in the 1980s.

159

Its success in clinical trials meant that the procedure was soon considered an

160

important component of the standard of care for newly diagnosed patients with MM

161

aged up to 65–70 years without significant comorbidities.32

162

A study comparing conventional treatments (8 cycles of RVD, plus stem cell

163

mobilization with high-dose cyclophosphamide and granulocyte colony-stimulating

164

factor after 3 cycles of RVD) with RVD treatment and ASCT (3 induction cycles of

165

RVD, followed by stem cell collection, and then ASCT with melphalan, followed by Page 9

Mikhael

166

2 cycles of RVD) reported a median PFS of 50 months in the transplant arm,

167

compared with 36 months in the RVD arm.10 The upfront trial of RVD versus RVD

168

plus ASCT demonstrated the continued value of ASCT in frontline therapy.33

169

However, it also speaks to the value of ASCT in relapsed disease, as 79% of

170

patients in the RVD alone arm had ASCT at first relapse.10 Salvage ASCT has now

171

been employed in later relapse, as a number of retrospective studies have also

172

demonstrated the efficacy of salvage ASCT following reinduction therapy in patients

173

with MM who have relapsed after a prior ASCT. The most important factor predicting

174

PFS after salvage ASCT is the duration of remission after initial ASCT, with those

175

patients with PFS of ≥ 18 months after their first autotransplant most likely to

176

benefit.34 Although recent guidelines from the International Myeloma Working Group

177

recommend that salvage ASCT is considered for all eligible patients,35 the procedure

178

is an option suitable only for a small minority. Many patients with MM are not

179

considered candidates due to their age and weakened state, arising as a

180

consequence of comorbidities, organ dysfunctions, and limitations in mental/mobility

181

functions, which would not enable them to withstand the procedure.20

182

Selinexor

183

Selinexor is a first-in-class selective inhibitor of nuclear export36 that provides an

184

additional therapeutic option for those with triple refractory disease; indeed, it has

185

been studied in patients with ‘quad- and penta-refractory disease’ by virtue of their

186

exposure to and becoming refractory to the 5 key agents used in this patient

187

population (bortezomib, lenalidomide, carfilzomib, pomalidomide, and

188

daratumumab). Selinexor is currently being evaluated as a component of

189

combination regimens in a number of phase II and III clinical trials in patients with

Page 10

Mikhael

190

RRMM.36 In the STORM (Selinexor Treatment of Refractory Myeloma) study

191

(NCT02336815), selinexor is being used in combination with low-dose

192

dexamethasone. The first part of this phase II trial evaluated selinexor and

193

dexamethasone in patients with MM refractory to bortezomib, carfilzomib,

194

lenalidomide, and pomalidomide, along with a small subset of patients with penta-

195

refractory disease, who were also refractory to an anti-CD38 antibody; indeed, this

196

was a very heavily pretreated population of patients with a median of 7 prior lines of

197

therapy.37 The ORR was 21% in patients with quad-refractory MM, and 20% for

198

patients with penta-refractory MM. Of note, the ORR was 35% among patients with

199

high-risk cytogenetics, including t(4;14), t(14;16), and del(17p). The median duration

200

of response was 5 months, and 65% of responding patients were alive at 12 months.

201

The most common grade ≥ 3 AEs were thrombocytopenia, anemia, neutropenia,

202

hyponatremia, leukopenia, and fatigue. Dose interruptions for AEs occurred in

203

41 patients, dose reductions occurred in 29 patients, and treatment discontinuation

204

occurred in 14 patients.37

205

Part 2 of STORM focused specifically on patients with penta-refractory MM, and

206

included many patients with rapidly progressive disease, with nearly 50% having

207

high-risk myeloma.38 In this context, selinexor in combination with low-dose

208

dexamethasone was highly active in this population, with an ORR of 26.2%.38

209

Treatment responses showed depth with a fast onset of action (within the first cycle

210

in most patients), with 2 patients achieving stringent CRs, both of whom were

211

minimal residual disease (MRD) negative.38, 39 Median OS was 8.6 months.38, 39 No

212

major organ toxicity was observed, and AEs were typically transient and

213

reversible.38, 39 Supportive care was important in reducing the severity of these

214

toxicities.38 In July 2019, selinexor was granted accelerated approval by the FDA for Page 11

Mikhael

215

adult patients with RRMM who have received at least four prior therapies and whose

216

disease is refractory to at least two proteasome inhibitors, at least two

217

immunomodulatory agents, and an anti-CD38 monoclonal antibody.40

218

The ongoing phase Ib/II STOMP (Selinexor and backbone Treatments Of Multiple

219

myeloma Patients) studies are evaluating selinexor and low-dose dexamethasone in

220

combination with a number of standard approved therapies including lenalidomide,

221

pomalidomide, bortezomib, carfilzomib, and daratumumab, and have generated

222

promising findings to date.41-43

223

Other Novel Approaches

224

Key clinical trial data for novel therapies are summarized in Table 1.

225

CAR T Cell Therapy

226

T cells can be genetically modified to express CARs, which are fusion proteins that

227

have an antigen recognition region and a co-stimulation domain.44 In MM, CAR T cell

228

therapies have shown clinical activity of up to 90–100% with a manageable safety

229

profile.44 Of note, bb2121 has shown promising efficacy at dose levels of

230

≥ 150×106 CAR T cells in 33 patients with RRMM who had received at least 3 prior

231

lines of therapy, including a proteasome inhibitor and an immunomodulatory agent,45

232

with a median PFS of 11.8 months. In total, 15 patients had a CR (45%)45 and

233

25 patients (76%) had cytokine release syndrome, which was of grade 3 in 2 patients

234

(6%).45 Cytokine release syndrome is the common toxicity associated with CAR T

235

cells, and is caused by the release of cytokines by the infused T cells.46 Built along

236

the bb2121 platform is bb21217, intended to have improved cell persistence. Early

237

efficacy results show 6 of 7 patients (median of 9 prior lines of therapy) Page 12

Mikhael

238

demonstrating clinical responses at a dose of 150x106 CAR T cells.47

239

In a heavily pretreated MM patient population that had failed IMiDs, proteasome

240

inhibitors and daratumumab, an overall response rate of 83% (5/6 patients) was

241

shown with P-BCMA-1.48 Only 1 patient developed cytokine release syndrome, of

242

limited severity (grade 2).48 The lack of significant toxicity observed with P-BCMA-1

243

is favorable compared to other CAR T cell therapies.48

244

Venetoclax

245

Venetoclax is a potent, selective, orally bioavailable small-molecule inhibitor of B cell

246

lymphoma (BCL)-2.36 Venetoclax has been investigated alone and in combination

247

with other therapies for the treatment of RRMM.49-51 Venetoclax monotherapy has an

248

acceptable safety profile and clear anti-myeloma activity in patients with RRMM,

249

primarily with t(11;14) having high BCL-2, low BCL-extra large, and low myeloid

250

leukemia cell differentiation protein-1 expression levels.49, 50 As part of a combination

251

regimen with bortezomib and dexamethasone, venetoclax has also demonstrated

252

efficacy in RRMM in a randomized phase III trial of venetoclax-bortezomib-

253

dexamethasone versus bortezomib-dexamethasone. Results revealed an improved

254

ORR of 82% and 68% and prolonged PFS of 22.4 months and 11.5 months,

255

respectively.52 However, there was a concerning safety signal with increased deaths

256

in the venetoclax-bortezomib-dexamethasone arm due to infections.52 The U.S. Food

257

and Drug Administration (FDA) placed a partial hold on trials with this agent in

258

myeloma and recommended that all patients with MM on this agent must be on

259

antibiotic prophylaxis. Subsequently, this partial hold has been lifted upon revisions

260

to the study protocol. It remains unclear what role this agent may play in myeloma,

261

but venetoclax could be a niche agent for patients with t(11;14) mutations, providing Page 13

Mikhael

262

clinicians with a means to identify patients likely to respond to certain treatments.52

263

This is important in the context of triple-class RRMM, as clinicians can be unsure as

264

to which therapies are the most appropriate for such a heavily pretreated population

265

that have failed standard MM therapies.

266

Programmed Cell Death Protein-1 Antibodies

267

Upregulation of the programmed cell death protein 1 (PD-1) pathway in MM prevents

268

the activation of antitumour T cell populations and can contribute to immune escape

269

by the tumour, enabling its proliferation.53 Pembrolizumab is an antibody that targets

270

PD-1, restoring the capacity of the immune system to perform cytotoxic T cell killing

271

of malignant plasma cells.53 Interim analysis from a phase III trial of pomalidomide

272

and dexamethasone with or without pembrolizumab in 249 RRMM patients with ≥2

273

prior lines of therapy showed an unfavourable benefit–risk profile for the triple

274

combination.54Median PFS was 5.6 months in the pembrolizumab plus

275

pomalidomide and dexamethasone group and 8.4 months in the pomalidomide and

276

dexamethasone-only group.54 Serious AEs (SAEs) occurred in 46% (56/121) of

277

patients in the pomalidomide and dexamethasone group, compared with 63%

278

(75/120) in the pembrolizumab plus pomalidomide and dexamethasone group.54

279

There were 4 (3%) treatment-related deaths with pembrolizumab plus pomalidomide

280

and dexamethasone.54 In the pomalidomide and dexamethasone-only group, no

281

treatment-related deaths were reported.54 Although these patients were not an

282

exclusively triple-class population, this study suggests that pembrolizumab does not

283

confer a survival benefit in RRMM.

Page 14

Mikhael

284

B Cell Maturation Antigen Antibodies

285

B cell maturation antigen (BCMA) is a cell surface receptor in the tumor necrosis

286

factor superfamily.44 Expression of BCMA is limited to B cells in advanced stages of

287

differentiation, and is required for the survival of long-lived plasma cells.44 Because

288

BCMA is expressed at significantly higher levels in all of the patient’s MM cells but

289

not on other normal tissues except normal plasma cells,44 it was identified as a target

290

for MM treatment. GSK2857916 is a humanized immunoglobulin G1 monoclonal

291

antibody with high affinity to BCMA conjugated to a microtubule-disrupting toxin

292

(monomethyl auristatin F). A phase I trial investigating monotherapy with the

293

antibody demonstrated a 38.5%% ORR and a median PFS of 6.2 months in those

294

who were refractory to daratumumab, a proteasome inhibitor and an

295

immunomodulator.55

296

Treatments Utilizing Bispecific T-cell Engager Technology

297

Bispecific T-cell Engagers (‘bispecifics’) contain 2 single-chain variable fragments

298

that are connected by a linker molecule. To redirect anticancer immunity, bispecifics

299

bind a T cell-specific antigen, often CD3, with 1 fragment, and a cancer-specific

300

epitope with the other, thus providing a platform to facilitate interaction between

301

effector and cancer cells.56 These may be particularly useful as the same product

302

can be given to all patients, whereas CAR T cell therapy is based on the patients’

303

own cells.57 The CD3-CD19 bispecific blinatumomab was granted FDA approval

304

based on the results of a phase II study that reported a 43% CR in patients with

305

relapsed or refractory B-cell precursor acute lymphoblastic leukemia.58 In RRMM,

306

preliminary results from the first-in-human study, in patients who had progression

307

after ≥2 lines of therapy (PI and IMiD), of the bispecific antibody construct AMG 420

Page 15

Mikhael

308

showed promising evidence of activity, with 13/42 responders.59 Of these, 3 were

309

CRs and 2 were PRs.59 The median time to any response was 1 month.59 Safety

310

was comparable to other immunotherapies, with SAEs in 50% of patients.59

311

Treatment-related SAEs included grade 3 polyneuropathies (n = 2) and edema (n =

312

1), both requiring hospitalization.59 Five MRD-negative stringent CRs were observed

313

in patients receiving AMG 420 400 µg/day.59

314

Other molecules are also being evaluated in RRMM, including inhibitors of murine

315

double minute 2 (MDM2), kinase, or bromodomain, along with other drug classes

316

that can induce apoptosis in MM cells.36

Page 16

Mikhael

317

Table 1. Novel approaches to triple-class RRMM

Name of agent 45

bb2121

bb21217

47

48

P-BCMA-101

Mechanism of action

Stage of development

Study number

Study population

Key efficacy results

Key safety results

CAR T cell

Phase I

NCT02658929

≥3 prior lines of therapy, including a PI and an IMiD, N = 33

ORR was 85%, CR in 15/33 (45%) of which 6 have since relapsed. Median PFS was 11.8 months (95% CI 6.2–17.8)

25/33 (76%) developed cytokine release syndrome, neurologic toxic effects in 14/33 (42%)

CAR T cell

Phase I

NCT03274219

≥3 prior lines of therapy, including a PI and an IMiD, N=8

6/7 demonstrated clinical response (1 sCR, 3 VGPR, 2 PR)

5/8 developed cytokine release syndrome

CAR T cell

Phase I

NCT03288493

≥3 prior lines of therapy, including a PI and an IMiD, N = 12

ORR was 83%

One patient has developed cytokine release syndrome (8%), most comment SAEs were cytopenia and febrile neutropenia

Page 17

Mikhael

Venetoclax (with bortezomib and 52 dexamethasone)

BCL-2 inhibitor

Phase III

NCT02755597

1–3 prior lines of therapy, N = 291

Mean PFS in the ventoclax arm was 22.4 months versus 11.5 months in the placebo arm, ORR was 82% vs 68%

41/194 (21.1%) deaths in the venetoclax arm, 11/97 (11.3%) in the placebo arm, severe, grade 3–5 toxicity and SAE rate were similar between the two arms.

Pembrolizumab (with pomalidomide and 54 dexamethasone)

Anti-PD-1 antibody

Phase III

NCT02576977

≥2 prior lines of therapy, N = 249

Median PFS in pembrolizumab arm was 5.6 months versus 8.4 months in the pomalidomide and dexamethasoneonly arm

4 deaths (3%) in the pembrolizumab arm. SAEs occurred in 75/120 (63%) in the pembrolizumab arm versus 56/121 (46%) in the pomalidomide and dexamethasone-only arm

BCMA

Phase I

NCT02064387

Prior therapy with an alkylator, PI and IMiD, N = 35

21/35 (60%) achieved a PR or better, including 2 sCR, and 3 CR. Median PFS was 12 months

Most commonly reported AEs were cough, increased aspartate aminotransferase and nausea (all grade 1 or 2). Corneal events and thrombocytopenia were manageable

Bispecific T-cell engager

Phase I

NCT02514239

Progression after ≥2 lines (incl PI and IMiD), N=42

13/42 responders including 6 sCRs, 3 CRs, 2 VGPR, 2 PRs. Median time to any response was 1 month

Treatment-related SAEs included 2 grade 3 polyneuropathy and 1 edema. Grade 2–3 cytokine release syndrome seen in 3 patients

GSK2857916

59

AMG 420

55

Page 18

Mikhael

318

BCL-2, B cell lymphoma 2; CAR, chimeric antigen receptor; CI, confidence interval; CR, complete response; IMiD, immunomodulatory agent; ORR, objective response rate;

319

PD-1, programmed cell death protein-1; PFS, progression-free survival; PI, proteasome inhibitor; PR, partial response; SAE, serious adverse event; sCR, stringent complete

320

response; VGPR, very good partial response

Page 19

Mikhael

321

Conclusion and Recommendations

322

In 2015, therapeutic options for MM expanded further when daratumumab and

323

elotuzumab were approved by the FDA for MM. Although monoclonal antibodies are

324

now of paramount importance in the context of MM care, their growing prominence is

325

coupled with the emergence of a new subset of patients who are refractory to

326

monoclonal antibodies, PIs, and IMiDs. The development and further refinement of

327

novel treatments for this subset of patients in the coming years should be considered

328

a key priority in myeloma. Current approaches to the treatment of triple-refractory

329

disease include conventional chemotherapy and salvage ASCT. It is anticipated that

330

in the near future, additional agents will be available for triple refractory disease,

331

namely selinexor, CAR T cell therapy, and next-generation monoclonal antibodies.

332

Therapeutic interventions for MM must be considered in the context of a supportive

333

care strategy that looks to improve patients’ quality of life and to help achieve better

334

treatment outcomes.60 Patients with RRMM are particularly vulnerable due to prior

335

exposure to chemotherapy, myelosuppression, long-term exposure to

336

corticosteroids, and impaired organ function.13 Supportive care must encompass

337

strategies to manage broader elements of MM, such as bone disease and spinal

338

cord compression, anemia, bone marrow failure and infections, and renal failure,33, 60

339

as well as management of treatment-related AEs.61

340

An understanding of disease-related factors, such as cytogenic abnormalities and

341

prognostic markers (eg, MRD),62, 63 and patient-related factors, such as renal

342

insufficiency, hepatic impairment, and comorbidities,62 is critical for evaluating

343

appropriate therapeutic options for patients with triple-class refractory MM.

344

Treatment-related factors, including prior drug exposures, longevity of remission, and Page 20

Mikhael

345

treatment-related toxicities,62 must also be considered. Further research is required

346

as part of continued efforts to identify and validate predictive biomarkers in MM that

347

can be used to guide treatment selection and to evaluate the efficacy of novel

348

therapies.64

Page 21

Mikhael

349

Disclosure

350

J.M. has acted as a consultant for AbbVie, Amgen, Celgene, Karyopharm

351

Therapeutics, Sanofi and Takeda. He has also received research funding from

352

AbbVie, Celgene, and Sanofi.

Page 22

Mikhael

353

Acknowledgments

354

The author acknowledges Hannah Burke, BSc (Core, London) for providing medical

355

writing support, and Rachael Cazaly, BSc (Core, London) for providing editorial

356

support, which was funded by Karyopharm Therapeutics and complied with Good

357

Publication Practice guidelines (Link). Karyopharm Therapeutics reviewed the

358

manuscript. However, the author is fully responsible for the opinions, conclusions,

359

and data interpretation presented in this manuscript.

Page 23

Mikhael

360

References

361

1.

Noll JE, Williams SA, Tong CM, et al. Myeloma plasma cells alter the bone

362

marrow microenvironment by stimulating the proliferation of mesenchymal

363

stromal cells. Haematologica 2014;99:163-171.

364

2.

Mikhael JR, Dingli D, Roy V, et al. Management of newly diagnosed

365

symptomatic multiple myeloma: updated Mayo Stratification of Myeloma and

366

Risk-Adapted Therapy (mSMART) consensus guidelines 2013. Mayo Clin

367

Proc 2013;88:360-376.

368

3.

Gay F, Engelhardt M, Terpos E, et al. From transplant to novel cellular

369

therapies in multiple myeloma: European Myeloma Network guidelines and

370

future perspectives. Haematologica 2018;103:197-211.

371

4.

Landgren O, Iskander K. Modern multiple myeloma therapy: deep, sustained

372

treatment response and good clinical outcomes. J Intern Med 2017;281:365-

373

382.

374

5.

Kumar SK, Callander NS, Alsina M, et al. Multiple Myeloma, Version 3.2017,

375

NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw

376

2017;15:230-269.

377

6.

Varga C, Maglio M, Ghobrial IM, Richardson PG. Current use of monoclonal

378

antibodies in the treatment of multiple myeloma. Br J Haematol 2018;181:447-

379

459.

Page 24

Mikhael

380

7.

Mahindra A, Laubach J, Raje N, Munshi N, Richardson PG, Anderson K.

381

Latest advances and current challenges in the treatment of multiple myeloma.

382

Nat Rev Clin Oncol 2012;9:135-143.

383

8.

384

385

Rajkumar SV. Multiple myeloma: 2016 update on diagnosis, risk-stratification, and management. Am J Hematol 2016;91:719-734.

9.

Palma BD, Prezioso L, Accardi F, et al. Addition of bortezomib to high dose

386

melphalan as conditioning regimen for autologous stem cell transplantation

387

improves the response rate in newly diagnosed multiple myeloma patients.

388

Blood 2016;128:4647.

389

10.

Attal M, Lauwers-Cances V, Hulin C, et al. Lenalidomide, bortezomib, and

390

dexamethasone with transplantation for myeloma. N Engl J Med

391

2017;376:1311-1320.

392

11.

Kumar SK, Dispenzieri A, Lacy MQ, et al. Continued improvement in survival

393

in multiple myeloma: changes in early mortality and outcomes in older

394

patients. Leukemia 2014;28:1122-1128.

395

12.

396

397 398

Fonseca R, Abouzaid S, Bonafede M, et al. Trends in overall survival and costs of multiple myeloma, 2000-2014. Leukemia 2017;31:1915-1921.

13.

Sonneveld P, Broijl A. Treatment of relapsed and refractory multiple myeloma. Haematologica 2016;101:396-406.

Page 25

Mikhael

399

14.

Usmani S, Ahmadi T, Ng Y, et al. Analysis of real-world data on overall

400

survival in multiple myeloma patients with >/=3 prior lines of therapy including

401

a proteasome inhibitor (PI) and an immunomodulatory drug (IMiD), or double

402

refractory to a PI and an IMiD. Oncologist 2016;21:1355-1361.

403

15.

Kumar SK, Dimopoulos MA, Kastritis E, et al. Natural history of relapsed

404

myeloma, refractory to immunomodulatory drugs and proteasome inhibitors: a

405

multicenter IMWG study. Leukemia 2017;31:2443-2448.

406

16.

Ghandhi U, Lakshman A, Gahvari Z, et al. Natural history of patients with

407

multiple myeloma refractory to CD38-targeted monoclonal aantibody-based

408

treatment. ASH 2018. Oral presentation 3233. Vol Poster number 3233. San

409

Diego, CA.

410

17.

Dimopoulos MA, Richardson PG, Moreau P, Anderson KC. Current treatment

411

landscape for relapsed and/or refractory multiple myeloma. Nat Rev Clin

412

Oncol 2015;12:42-54.

413

18.

414

415

refractory multiple myeloma. Clin Cancer Res 2011;17:1264-1277.

19.

416

417 418

Lonial S, Mitsiades CS, Richardson PG. Treatment options for relapsed and

Mikhael JR. A practical approach to relapsed multiple myeloma. Hematology Am Soc Hematol Educ Program 2014;2014:262-267.

20.

Palumbo A, Rajkumar SV, San Miguel JF, et al. International Myeloma Working Group consensus statement for the management, treatment, and

Page 26

Mikhael

419

supportive care of patients with myeloma not eligible for standard autologous

420

stem-cell transplantation. J Clin Oncol 2014;32:587-600.

421

21.

Toocheck C, Pinkhas D. Treatment of relapsed multiple myeloma complicated

422

by cardiac extramedullary plasmacytoma with D-PACE chemotherapy. BMJ

423

Case Rep 2018:pii: bcr-2017-223611.

424

22.

Nikonova A, Caplan SN, Shamy A, Gyger M. High-dose cyclophosphamide in

425

highly refractory multiple myeloma patients as a bridge to further novel

426

therapies. Am Soc Hematology 2016;128:5676.

427

23.

428

429

Michael M, Bruns I, Bolke E, et al. Bendamustine in patients with relapsed or refractory multiple myeloma. Eur J Med Res 2010;15:13-19.

24.

Gandhi UH, Cornell RF, Lakshman A, et al. Outcomes of patients with

430

multiple myeloma refractory to CD38-targeted monoclonal antibody therapy.

431

Leukemia 2019.

432

25.

Dingli D, Ailawadhi S, Bergsagel PL, et al. Therapy for relapsed multiple

433

myeloma: guidelines from the mayo stratification for myeloma and risk-

434

adapted therapy. Mayo Clin Proc 2017;92:578-598.

435 436

26.

Deleu S, Menu E, Valckenborgh EV, et al. Histone deacetylase inhibitors in multiple myeloma. Hematology Rev 2009;1:e9.

Page 27

Mikhael

437

27.

438

439

Yee AJ, Raje NS. Panobinostat and multiple myeloma in 2018. Oncologist 2018;23:516-517.

28.

San-Miguel JF, Hungria VT, Yoon SS, et al. Panobinostat plus bortezomib

440

and dexamethasone versus placebo plus bortezomib and dexamethasone in

441

patients with relapsed or relapsed and refractory multiple myeloma: a

442

multicentre, randomised, double-blind phase 3 trial. Lancet Oncol

443

2014;15:1195-1206.

444

29.

Richardson PG, Hungria VTM, Yoon S-S, et al. Panobinostat plus bortezomib

445

and dexamethasone in previously treated multiple myeloma: outcomes by

446

prior treatment. Blood 2016;127:713-721.

447

30.

448

449

Greig SL. Panobinostat: a review in relapsed or refractory multiple myeloma. Target Oncol 2016;11:107-114.

31.

Vij R, Chen C, Yasenchak C, Davis C, Kuter D. Treatment Sequencing in

450

Patients with Relapsed/Refractory Multiple Myeloma after Daratumumab

451

Treatment: Real-World Findings from a Pooled Data Analysis of Preamble

452

and the Mckesson Electronic Medical Record Database. Blood

453

2018;132:3284.

454

32.

Cook G, Ashcroft AJ, Cairns DA, et al. The effect of salvage autologous stem-

455

cell transplantation on overall survival in patients with relapsed multiple

456

myeloma (final results from BSBMT/UKMF Myeloma X Relapse [Intensive]): a

457

randomised, open-label, phase 3 trial. Lancet Haematol 2016;3:e340-351. Page 28

Mikhael

458

33.

Moreau P, San Miguel J, Sonneveld P, et al. Multiple myeloma: ESMO clinical

459

practice guidelines for diagnosis, treatment and follow-up. Ann Oncol

460

2017;28:iv52-iv61.

461

34.

Chim CS, Kumar SK, Orlowski RZ, et al. Management of relapsed and

462

refractory multiple myeloma: novel agents, antibodies, immunotherapies and

463

beyond. Leukemia 2018;32:252-262.

464

35.

Giralt S, Garderet L, Durie B, et al. American Society of Blood and Marrow

465

Transplantation, European Society of Blood and Marrow Transplantation,

466

Blood and Marrow Transplant Clinical Trials Network, and International

467

Myeloma Working Group Consensus Conference on Salvage Hematopoietic

468

Cell Transplantation in Patients with Relapsed Multiple Myeloma. Biol Blood

469

Marrow Transplant 2015;21:2039-2051.

470

36.

Abramson HN. The multiple myeloma drug pipeline-2018: a review of small

471

molecules and their therapeutic targets. Clin Lymphoma Myeloma Leuk

472

2018;18:611-627.

473

37.

Vogl DT, Dingli D, Cornell RF, et al. Selective inhibition of nuclear export with

474

oral selinexor for treatment of relapsed or refractory multiple myeloma. J Clin

475

Oncol 2018;36:859-866.

476 477

38.

Chari A, Vogl D, Dimopoulos M, et al. Results of the pivotal STORM study (Part 2) in penta-refractory multiple myeloma (MM): deep and durable

Page 29

Mikhael

478

responses with oral selinexor plus low dose dexamethasone in patients with

479

penta-refractory MM. Blood 2018;132 Suppl 1:598.

480

39.

Jagannath S, Vogl DT, Dimopoulos M-A, et al. Phase 2b results of the

481

STORM study: oral selinexor plus low dose dexamethasone (Sd) in patients

482

with penta-refractory myeloma (penta-MM). Clin Lymphoma Myeloma Leuk

483

2018;18:S249-S250.

484

40.

U.S. Food and Drug Administration. FDA grants accelerated approval to

485

selinexor for multiple myeloma. https://www.fda.gov/drugs/resources-

486

information-approved-drugs/fda-grants-accelerated-approval-selinexor-

487

multiple-myeloma; 2019 Accessed July 2019.

488

41.

Chen C, Kotb R, Sebag M, et al. Selinexor shows synergy in combination with

489

pomalidomide and low dose dexamethasone in patients with relapsed /

490

refractory multiple myeloma. Blood 2016;128:3330-3330.

491

42.

Bahlis NJ, Sutherland H, White D, et al. Selinexor plus low-dose bortezomib

492

and dexamethasone for patients with relapsed or refractory multiple myeloma.

493

Blood 2018;132:2546-2554.

494

43.

ClinicalTrials.gov. Selinexor and backbone Treatments Of Multiple myeloma

495

Patients (STOMP). https://clinicaltrials.gov/ct2/show/NCT02343042; 2015

496

Accessed June 7, 2019.

Page 30

Mikhael

497

44.

Cho SF, Anderson KC, Tai YT. Targeting B cell maturation antigen (BCMA) in

498

multiple myeloma: potential uses of BCMA-based immunotherapy. Front

499

Immunol 2018;9:1821.

500

45.

Raje N, Berdeja J, Lin Y, et al. Anti-BCMA CAR T-Cell Therapy bb2121 in

501

Relapsed or Refractory Multiple Myeloma. N Engl J Med 2019;380:1726-

502

1737.

503

46.

504

505

Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 2016;127:3321-3330.

47.

Shah N, Alsina M, Siegal D, et al. Initial Results from a Phase 1 Clinical Study

506

of bb21217, a Next-Generation Anti Bcma CAR T Therapy. Blood

507

2018;132:488.

508

48.

Gregory T, Cohen A, Costello S, et al. Efficacy and Safety of P-Bcma-101

509

CAR-T Cells in Patients with Relapsed/Refractory (r/r) Multiple Myeloma

510

(MM). Blood 2018;132:1012.

511

49.

Kumar S, Vij R, Kaufman JL, et al. Venetoclax monotherapy for

512

relapsed/refractory multiple myeloma: safety and efficacy results from a phase

513

I study. Blood 2016;128:488-488.

514

50.

Kumar S, Kaufman JL, Gasparetto C, et al. Efficacy of venetoclax as targeted

515

therapy for relapsed/refractory t(11;14) multiple myeloma. Blood

516

2017;130:2401-2409.

Page 31

Mikhael

517

51.

Moreau P, Chanan-Khan AA, Roberts AW, et al. Venetoclax combined with

518

bortezomib and dexamethasone for patients with relapsed/refractory multiple

519

myeloma. Blood 2016;128:975-975.

520

52.

U.S. Food and Drug Administration. FDA warns about the risks associated

521

with the investigational use of Venclexta in Multiple Myeloma.

522

https://www.fda.gov/Drugs/DrugSafety/ucm634120.htm; 2019 Accessed June

523

7, 2019.

524

53.

525

526

Rosenblatt J, Avigan D. Targeting the PD-1/PD-L1 axis in multiple myeloma: a dream or a reality? Blood 2017;129:275-279.

54.

Mateos MV, Blacklock H, Schjesvold F, et al. Pembrolizumab plus

527

pomalidomide and dexamethasone for patients with relapsed or refractory

528

multiple myeloma (KEYNOTE-183): a randomised, open-label, phase 3 trial.

529

Lancet Haematol 2019.

530

55.

Trudel S, Lendvai N, Popat R, et al. Antibody-drug conjugate, GSK2857916,

531

in relapsed/refractory multiple myeloma: an update on safety and efficacy

532

from dose expansion phase I study. Blood Cancer J 2019;9:37.

533 534

56.

Bianchi G, Richardson PG, Anderson KC. Promising therapies in multiple myeloma. Blood 2015;126:300-310.

Page 32

Mikhael

535

57.

Gohil SH, Paredes-Moscosso SR, Harrasser M, et al. An ROR1 bi-specific T-

536

cell engager provides effective targeting and cytotoxicity against a range of

537

solid tumors. Oncoimmunology 2017;6:e1326437.

538

58.

Topp MS, Gokbuget N, Stein AS, et al. Safety and activity of blinatumomab

539

for adult patients with relapsed or refractory B-precursor acute lymphoblastic

540

leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol

541

2015;16:57-66.

542

59.

Topp M, Duell J, Zugmaier G, et al. Evaluation of AMG 420, an anti-BCMA

543

bispecific T-cell engager (BiTE) immunotherapy, in R/R multiple myeloma

544

(MM) patients: Updated results of a first-in-human (FIH) phase I dose

545

escalation study. ASCO. Chicago, IL2019.

546

60.

547

548

Snowden JA, Ahmedzai SH, Ashcroft J, et al. Guidelines for supportive care in multiple myeloma 2011. Br J Haematol 2011;154:76-103.

61.

Kurtin SE, Bilotti E. Novel agents for the treatment of multiple myeloma:

549

proteasome inhibitors and immunomodulatory agents. J Adv Pract Oncol

550

2013;4:307-321.

551 552

62.

Nooka AK, Kastritis E, Dimopoulos MA, Lonial S. Treatment options for relapsed and refractory multiple myeloma. Blood 2015;125:3085-3099.

Page 33

Mikhael

553

63.

Perrot A, Lauwers-Cances V, Corre J, et al. Minimal residual disease

554

negativity using deep sequencing is a major prognostic factor in multiple

555

myeloma. Blood 2018;132:2456-2464.

556

64.

Nijhof IS, van de Donk N, Zweegman S, Lokhorst HM. Current and new

557

therapeutic strategies for relapsed and refractory multiple myeloma: an

558

update. Drugs 2018;78:19-37.

559

Page 34