AngioDynamics to Present at the Needham Healthcare Conference

On April 1, 2019 AngioDynamics, Inc. (NASDAQ: ANGO), a leading provider of innovative, minimally invasive medical devices for vascular access, peripheral vascular disease, and oncology, reported that Michael C. Greiner, Executive Vice President and Chief Financial Officer, will present at the 18th Annual Needham Healthcare Conference at 1:30 p.m. ET on Tuesday, April 9, 2019 in New York, NY (Press release, AngioDynamics, APR 1, 2019, View Source [SID1234534852]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

A live webcast of the presentation will be accessible through the "Investors" section of the Company’s website at www.angiodynamics.com and will be available for replay following the event.

Development of the engExTM Platform for Engineered Exosomes and Therapeutic Potential of Codiak’s exoSTING Highlighted at the 2019 American Association for Cancer Research Annual Meeting

On April 1, 2019 Codiak BioSciences, Inc., a leading exosome therapeutics company, reported data supporting the therapeutic potential of the engExTM Platform for engineering exosomes and its exoSTING product candidate as highlighted at the 2019 American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting, held March 31 through April 3 in Atlanta, Georgia (Press release, Codiak Biosciences, APR 1, 2019, View Source [SID1234534851]). Using its engEx Platform, Codiak engineers exosomes — natural intercellular messengers — to have precise and intentionally chosen properties, to incorporate various types of biologically active molecules, and to be directed to specific cell types and tissues. exoSTING is Codiak’s precision engineered exosome therapeutic candidate loaded with a potent small molecule STING (stimulator of interferon genes) agonist. Codiak is conducting IND-enabling studies of exoSTING and plans to begin a Phase 1/2 trial in the immuno-oncology setting during the first half of 2020.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

exoSTING

exoSTING is one of Codiak’s lead exosome therapeutic candidates that specifically targets the STING pathway in antigen presenting cells (APCs). exoSTING has been evaluated in a variety of preclinical studies including syngeneic tumor models refractory to checkpoint therapy, the results of which were featured in a presentation during the meeting’s Cancer Vaccines and Intratumoral Immunomodulation Minisymposium. Key conclusions from the preclinical analysis highlight the unique profile of exoSTING to selectively activate the STING pathway in tumor-resident APCs, resulting in systemic anti-tumor immunity without toxic systemic cytokine elevation:

exoSTING demonstrated enhanced potency and selective activation in human peripheral blood mononuclear cells (PBMCs), including monocytes and myeloid dendritic cells, compared to free STING agonist.
Exosomes loaded with the novel exosome surface glycoprotein prostaglandin F2 receptor negative regulator (PTGFRN, formerly designated Protein X) demonstrated enhanced APC activation and improved potency in a B16F10 tumor model as compared to the free STING agonist.
When delivered intratumorally, exoSTING displayed tumor retained pharmacology, with enhanced anti-tumoral effect and reduced systemic inflammatory cytokine production compared to optimal doses of free STING.
exoSTING preserved tumor resident T-cells and APCs to induce better tumor antigen specific immune response.
exoSTING improved anti-tumor immunity compared to free STING agonist in primary subcutaneous B16F10 tumors as well as non-primary distant (lung) tumors, where complete responses were histologically confirmed. Complete responses were not seen with free STING.
This anti-tumor immunity mediated by exoSTING is dependent on CD8 T-cells.
engEx Platform

The potential of the engEx Platform to generate novel exosome therapeutic candidates was characterized in a poster during the Novel Screening and Delivery Technologies Session at AACR (Free AACR Whitepaper). With the engEx Platform, Codiak aims to functionalize the surface of its engineered exosomes with proteins and peptides– an important strategy to harness their therapeutic potential. Comparative proteomic analysis of stringently purified exosomes led to the identification of several highly enriched and unique proteins, including PTGFRN. Codiak has developed precision engineered exosome therapeutic candidates using PTGFRN as a scaffold to enable high-density surface display of structurally and biologically diverse proteins, including enzymes, antibodies, type I cytokines, and TNF superfamily members. When these proteins were genetically fused to PTGFRN and overexpressed in a producer cell, significantly higher transgene expression on secreted exosomes was achieved compared to previously published scaffolds. PTGFRN-mediated display of CD40L on engineered exosomes resulted in a 15-fold potency increase in B cell activation over recombinant CD40L. Furthermore, expression of CD40L redirected exosome uptake from phagocytic APCs to B cells, demonstrating exosome surface modifications can alter cellular tropism. Codiak exosomes with IL-12 tethered to the surface demonstrated superior tumor retention compared to free cytokine, resulting in robust anti-tumor activity in anti-PD-1 refractory B16F10 tumor models. Additionally, exosomes engineered to overexpress PTGFRN enhanced activity of exosome-mediated delivery of STING agonist, with expression levels positively correlating with IFNβ production and tumor growth inhibition

Sensei Biotherapeutics Presents First Preclinical Data on SNS-723 CAR-T and Long-Term Results on SNS-301 Data at the 2019 American Association for Cancer Research (AACR) Annual Meeting

On April 1, 2019 Sensei Biotherapeutics, Inc., a clinical-stage biopharmaceutical company developing precision immuno-oncology therapies, reported new data from two programs targeting a novel tumor specific embryonic antigen, human aspartate β-hydroxylase (ASPH), including preclinical data on SNS-723, a first-in-class CAR-T cell therapy demonstrating effective killing of tumor cells by a series of ASPH-specific CAR-Ts as well as additional data from its Phase 1 study on the long-term effects of SNS-301, a first-in-class cancer immunotherapy demonstrating that all patients experienced dose-dependent and durable ASPH-specific immune responses (Press release, Sensei Biotherapeutics, APR 1, 2019, View Source [SID1234534850]). These data were presented in two poster presentations at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting in Atlanta, Georgia from March 29 – April 3, 2019.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"We are excited to share these first preclinical data for SNS-723," said Hossein Ghanbari, Ph.D., Chief Scientific Officer of Sensei Biotherapeutics. "This novel CAR-T is designed to elicit tumor cell destruction where ASPH is expressed, which includes both solid tumors and hematological malignancies. Given that ASPH is not found on the surface of normal tissues at any detectable levels, we believe SNS-723 offers a substantial opportunity to improve on the safety profile of today’s approved cell therapies."

"These long-term data from our phase 1 study of SNS-301 are consistent with our earlier findings of strong dose-dependent APSH-specific immunogenicity in patients and provide additional insights into our Phase 2 dosing strategy," said Ildiko Csiki, M.D., Ph.D., Chief Medical Officer of Sensei Biotherapeutics. "Given the promise of targeting ASPH as a new strategy for treating cancer, we remain focused on initiating Phase 2 trials of SNS-301 in various hematological malignancies and solid tumors in 2019."

In a poster titled "CAR-T Cell Therapies Targeting Aspartyl β-hydroxylase (ASPH)," Sensei researchers examined multiple ASPH-specific CAR constructs for CAR expression, cell-killing efficacy and cytokine response. Key SNS-723 data (poster #2306) highlights included:

Significant expression of three different evaluated ASPH-specific CARs was observed in human T-cells.
Dose-dependent cell-killing of ASPH-expressing H460 lung carcinoma cells by two of the ASPH-specific CAR constructs was observed concomitant with cytokine release. In contrast, untransduced T-cells or T-cells transduced to express a non-relevant CAR did not kill target cells of or activate cytokine production.
CARs binding two different, non-overlapping epitopes on ASPH both displayed cell killing efficacy.
In a poster titled "Long-term Immunogenicity Results from a First-in-human Study Evaluating the Anti-ASPH Cancer Vaccine, SNS-301," Sensei researchers examined both short- and long-term immune responses to SNS-301. Previous data from the Phase 1 clinical trial of SNS-301 were presented in 2018, demonstrating rapid and significant antigen-specific B-cell and T-cell responses induced by SNS-301. In the extension of the Phase 1 study, the 12 enrolled patients received between 8 and 23 doses of the vaccine delivered via an intradermal injection every 21 days. The additional key SNS-301 data (poster #1454) highlights included:

All patients continued to experience long-term, dose-dependent and durable ASPH-specific immune responses, including B-cell, T-cell, and antibody responses.
A fluctuation in the peak immune responses following the first six cycles was seen, suggesting the possibility of immune fatigue and a potential benefit from increased spacing between boosting doses after the first 6 cycles.
Anti-phage antibody responses were generally much lower at the low- and mid-doses than the high dose. Based on the ratio of ASPH-specific immune response to anti-phage immune response, the mid-dose is the recommended Phase 2 dose.
Based on the data presented today, Sensei plans to initiate multiple multi-site Phase 2 clinical trials in mid-2019 to evaluate the immunogenicity and preliminary clinical activity of SNS-301 in various malignancies. Sensei also plans to continue preclinical development and begin IND-enabling studies for SNS-723 in the second half of 2019.

About SNS-723

SNS-723 is a first-in-class CAR-T cell therapy that is currently in preclinical development targeting human aspartate β-hydroxylase (ASPH), a cell surface enzyme that is normally expressed during fetal development. The recognition domain of the CAR is the scFv portion of a high affinity, fully human, anti-ASPH antibody. SNS-723 is designed to overcome one of the major hurdles in T-cell therapy, targeting T-cells to tumors in the absence of non-tolerable and/or off-target toxicities to essential tissues and organs. Experiments to further characterize ASPH-targeted CAR-T cells are ongoing with the goal of moving these promising therapeutics into clinic.

About SNS-301

SNS-301 is a first-in-class cancer immunotherapy targeting human aspartate β-hydroxylase (ASPH), a cell surface enzyme that is normally expressed during fetal development. Following fetal development, the protein is no longer expressed. Expression of ASPH is uniquely upregulated in more than 20 different cancer types and promotes cancer cell growth, cell motility and invasiveness. ASPH expression levels in various tumors are inversely correlated with tumor resistance and patient survival. Through enhanced antigen presentation and other engineered immunotherapeutic features, SNS-301 is designed to overcome self- tolerance and induce robust and durable ASPH-specific humoral and cellular immune responses. SNS-301 is paired with a companion diagnostic to select antigen-positive patients and is delivered intradermally for ease of administration.

Rgenix To Present Abstract on RGX-019 at the 2019 AACR Annual Meeting

On April 1, 2019 Rgenix, Inc., a clinical stage biopharmaceutical company developing first-in-class small molecule and antibody cancer therapeutics, reported that Isabel Kurth, Ph.D., Vice President of Research at Rgenix, will present an abstract about Rgenix’s RGX-019 program at the 2019 American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting (Press release, Rgenix, APR 1, 2019, View Source [SID1234534849]). The meeting is scheduled to take place Friday, March 29 through Wednesday, April 3 in Atlanta.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

The details of Rgenix’s presentation are as follows:


Event:
AACR Annual Meeting 2019

Date: April 3, 2019

Time: 8:00 A.M. – 12:00 P.M. EST

Description: Abstract LB-277/23, "Characterization of the anti-cancer and immunologic activity of RGX-019, a novel pre-clinical stage humanized monoclonal antibody targeting the MERTK receptor"

Location: Section 40, Georgia World Congress Center, 285 Andrew Young International Blvd
NW, Atlanta, GA 30313

Dr. Kurth will present data from pre-clinical research of RGX-019, a novel pre-clinical stage humanized monoclonal antibody targeting the MERTK receptor.

CELGENE RECEIVES THE CHMP’S POSITIVE OPINION FOR THE TRIPLETTE BASED ON REVLIMID ® (LENALIDOMIDE) AND IMNOVID ® (POMALIDOMIDE) IN COMBINATION WITH BORTEZOMIB AND DESAMETASONE IN MULTIPLE MYELOMA PATIENTS

On April 1, 2019 Celgene Corporation (NASDAQ: CELG) reported that the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) has expressed a positive opinion for the two triplets in basis of the IMiD of Celgene, REVLIMID (lenalidomide) and IMNOVID (pomalidomide) (Press release, Celgene, APR 1, 2019, View Source [SID1234534848]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

The CHMP recommended the approval of the extension of the lenalidomide indication as combination therapy with bortezomib and dexamethasone (RVd) for the treatment of adult patients with newly diagnosed multiple myeloma not eligible for transplantation.

The Committee also recommended the approval of pomalidomide in combination with bortezomib and dexamethasone (PVd) for the treatment of adult patients with multiple myeloma who have received at least one previous lenalidomide treatment.

The final decision of the European Commission, which generally conforms to the CHMP’s recommendations, is expected in about two months.

" The CHMP’s positive opinion regarding the RVd and PVd triplets represents an important extension of therapy options for patients with multiple myeloma, both newly diagnosed and relapsed / refractory after previous treatment lines, " commented Michele Cavo , Full Professor of Hematology, University of Bologna and Director of the "Seràgnoli" Hematology Institute, S. Orsola-Malpighi University Hospital, Bologna – In fact, RVd is configured as a more effective therapeutic alternative with respect to the current standards of first-line treatment of patients who are not eligible to receive an autologous transplant, to which it ensures a significant prolongation both of overall survival (whose median value was higher than 6 years in the pivotal study) that of disease-free progression survival (median value greater than 40 months). Instead, PVd represents one of the most effective combinations currently available for the rescue therapy of relapsed or refractory patients after previous exposure to lenalidomide, and in particular to those who have become refractory to it.» .

The CHMP’s positive opinion for lenalidomide was based on data from SWOG S0777, a phase 3 study that evaluated the triplet with lenalidomide, bortezomib and dexamethasone (RVd) in adult patients with previously untreated multiple myeloma, not immediately eligible for ASCT 1 . The results of the study showed statistically significant improvements both in terms of progression-free survival (PFS) and overall survival (OS) in patients treated with RVd compared to those treated with lenalidomide plus dexamethasone (Rd). The choice of first-line treatment is important 2since over time patients respond less and less to therapy, manifesting increasingly shorter periods of remission in subsequent treatment lines. 3

The CHMP’s positive opinion for PVd was based on data from OPTIMISMM, the first prospective phase 3 study that evaluated the use of the triplet with pomalidomide in patients previously treated with lenalidomide who in most cases (70%) were found refractory to it. 4 This patient population represents a growing unmet medical need for which new therapeutic options are needed.

The OPTIMISMM results showed a significantly higher progression-free survival in patients treated with PVd compared to patients in the Vd treatment arm.

" The positive opinions of the CHMP for our combinations of IMiD RVd and PVd represent wonderful news for patients with multiple myeloma living in Europe, " says Nadim Ahmed , President of the Hematology / Oncology Division of Celgene. " We now hope that the EMA will approve these therapeutic options by making them available to patients, as our goal is to improve their outcomes at different stages of the disease ."

Lenalidomide in combination with bortezomib and dexamethasone, as well as pomalidomide in combination with bortezomib and dexamethasone, are currently not approved in any country.

Information on Celgene immunomodulatory drugs

IMiD agents are small molecules available as an oral therapy for the treatment of certain hematological malignancies owned exclusively by Celgene. It is hypothesized that they have multiple mechanisms of action. It has been observed that T cell activation and proliferation, IL-2 protein proliferation and CD8 + effector T cell activity are observed to increase. They also influence the stimulation and expression of natural killer cells(NK) acting within the cellular environment with the aim of inducing the immune system to attack cancer cells and at the same time directly attack them. In addition to the immunomodulatory properties, a tumoricidal and antiangiogenic activity is also hypothesized. Celgene IMiDs represent a mainstay of research on multiple myeloma with a growing number of studies in combination therapies in different disease settings.

Information on multiple myeloma

Multiple myeloma is a potentially fatal haematological neoplasm, which is characterized by tumor proliferation and suppression of the immune system. 5.6 It is a rare but lethal disease: every year, in Europe, there are 42,000 diagnoses, with about 26,000 deaths due to the disease. 7 The typical course of multiple myeloma includes periods of symptomatic disease alternating with periods of remission and, with time, appearance of refractoriness (non-responsiveness). 8

Information on the SWOG study S0777

SWOG S0777 is a multicenter, phase 3, randomized open trial, aimed at evaluating the efficacy and safety of RVd compared to Rd in the treatment of patients with previously untreated multiple myeloma for whom an autologous transplant of stem cells (ASCT). 1

In this study, 525 patients with newly diagnosed multiple myeloma (NDM) were enrolled with symptomatic and measurable disease aged 18 years or older. Patients were randomized (1: 1) to receive initial lenalidomide treatment with bortezomib and dexamethasone (RVd group) or lenalidomide and dexamethasone therapy only (group Rd). Randomization was stratified based on the ISS stage ( International Staging System, I, II or III ) and intention to transplant (yes vs.no). The RVd regimen was administered in eight cycles lasting 21 days. Bortezomib was administered at a dose of 1.3 mg / m by IV route on days 1, 4, 8 and 11, in combination with oral lenalidomide 25 mg / day on days 1-14 plus oral dexamethasone at a dose of 20 mg / day on days 1, 2, 4, 5, 8, 9, 11 and 12. The Rd regimen was administered in six cycles lasting 28 days. The Rd standard regimen consisted of 25 mg / day of lenalidomide orally on days 1-21 plus 40 mg / day of dexamethasone orally on days 1, 8, 15 and 22. 1

The results of the SWOG S0777 1 study showed a significant improvement in median progression-free survival (mPFS) in patients treated with RVd vs. Rd (42 vs. 30 months; HR 0.76, 95% CI 0.62-0.94; P = 0.01). A significant improvement in median overall survival with respect to Rd was also observed with RVd (89 vs. 67 months; HR 0.72, 95% CI 0.56-0.94; P = 0.013). The overall response and complete response rates were superior in patients treated with RVd compared to Rd (global response: 82% RVd vs 72% Rd; complete response: 16% RVd vs. 8% Rd). The safety of RVd was also in line with the consolidated safety profiles of each drug in the triplet. 9.10

Upon completion of the induction, all patients received continuous maintenance therapy with lenalidomide orally at 25 mg once a day for 21 days plus oral dexamethasone at a dose of 40 mg once a day on days 1 , 8, 15 and 22 of each cycle lasting 28 days. 1

Information on the OPTIMISMM study

OPTIMISMM is the first phase 3 comparative study on the safety and efficacy of PVd vs. Vd, as an early therapy line in patients with relapsed and refractory multiple myeloma (with 1-3 previous therapeutic regimens) and previous exposure to lenalidomide, including patients refractory to it. 4

This international, multi-center, phase 3, randomized, open-label study involved 559 patients (281 patients in the PVd arm and 278 in the Vd arm) with previous demographic, basal and pathological features generally well balanced between the two treatment arms . The median number of previous treatment lines was two, while over one third of patients had received a previous treatment line (40% in both treatment arms). All patients had previously been treated with lenalidomide: most were refractory to lenalidomide (71% in the PVd vs. arm69% in the Vd arm); last treatment refractoriness was observed in 70% and 66% of cases respectively. The median follow-up was 16 months. 4

The results of the OPTIMISMM study showed a significantly higher PFS in patients treated with PVd compared to patients in the Vd arm (11.20 vs. 7.10 months [P = <0.0001, HR 0.61; 95% CI: (0.49-0.77)]), with a 39% reduction in the risk of disease progression or death in the PVd arm. In an exploratory analysis on the subgroup of patients undergoing a previous treatment line, the median progression-free survival (mPFS) was 20.73 months with PVd vs.11.63 months with Vd (HR 0.54; p = 0.0027). In these patients, the benefit of PVd was independent of refractoriness or non-refractory to previous lenalidomide therapy. The safety of PVd was in line with the established safety profiles of each drug in the triplet. 7.11

Patients were stratified according to the following criteria: age (≤75 years vs. > 75 years), number of previous therapeutic regimens for myeloma (1 vs. > 1) and β2-microglobulin levels (from <3.5 mg / L vs. ≥ 3.5 to ≤ 5.5 mg / L vs.> 5.5 mg / L). Patients were randomized 1: 1 to receive PVd or Vd. In 21-day cycles, patients received pomalidomide 4 mg / day on days 1-14 (PVd arm only); bortezomib 1.3 mg / m2 on days 1, 4, 8 and 11 of cycles 1-8 and on days 1 and 8 of cycle 9 and subsequent; and dexamethasone 20 mg / day (10 mg if age was> 75 years) on days when treatment with bortezomib was expected and on subsequent days. 4

Information on REVLIMID (lenalidomide)

Lenalidomide is approved in Europe as a monotherapy and is indicated for the maintenance therapy of adult patients with newly diagnosed multiple myeloma who have undergone an autologous stem cell transplant. Lenalidomide as a combination therapy is approved in Europe, the United States, Japan and about 25 other countries for the treatment of adult patients with multiple previously treated myeloma (MM) who are not eligible for transplantation. It is also approved in association with dexamethasone for the treatment of patients with MM who have received at least one previous therapy in almost 70 countries in Europe, the Americas, the Middle East and Asia

Lenalidomide is also approved in the United States, Canada, Switzerland, Australia, New Zealand and in several Latin American countries, as well as in Malaysia and Israel, for the treatment of transfusion-dependent anemia due to myelodysplastic syndromes (MDS) to low or intermediate-1 risk, associated with an isolated cytogenetic abnormality with deletion of 5q, with or without further cytogenetic abnormalities, as well as in Europe for the treatment of patients with transfusion-dependent anemia due to low or intermediate-1 myelodysplastic syndromes , associated with an isolated cytogenetic abnormality with deletion of 5q, in cases where other therapeutic options have proved insufficient or inadequate.

It is also approved in Europe and the United States for the treatment of patients with relapsed or refractory mantle lymphoma (MCL) after two previous therapies, one of which includes bortezomib. In Switzerland, Lenalidomide is indicated for the treatment of patients with relapsed or refractory MCL after previous therapy with bortezomib and chemotherapy / rituximab.

Lenalidomide is not indicated and is not recommended for the treatment of patients with chronic lymphocytic leukemia (CLL) outside of controlled clinical studies.

About IMNOVID (pomalidomide)

Pomalidomide administered in combination with dexamethasone is indicated in the treatment of adult patients with relapsed and refractory multiple myeloma who have undergone at least two previous therapies, including both lenalidomide and bortezomib and with demonstrated progression of the disease during the last therapy. It belongs to the class of so-called immunomodulatory drugs (IMiD ).

Pomalidomide was initially approved in the United States in 2013 in association with dexamethasone for the treatment of patients with multiple myeloma who received at least two previous therapies including lenalidomide and a proteasome inhibitor and with demonstrated progression of the disease during or within 60 days of completion of the latest therapy.

In 2013 it was approved in the EU, in association with dexamethasone for the treatment of adult patients with relapsed and refractory multiple myeloma, who underwent at least two previous therapies, including both lenalidomide and bortezomib and with demonstrated progression of the disease during the last therapy.

It is also approved in a total of 66 countries in the world, including Australia, Canada, Japan and Switzerland, for use in association with dexamethasone for indications similar to those approved in the United States and the EU.