MorphoSys AG Announces Third Quarter 2018 Results

On November 5, 2018 MorphoSys AG (FSE: MOR; Prime Standard Segment, MDAX & TecDAX; NASDAQ: MOR) reported financial results for the third quarter and first nine months of 2018 (Press release, MorphoSys, NOV 5, 2018, View Source [SID1234530766]).

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 third quarter of 2018 was a productive one for MorphoSys, highlighted by very encouraging progress in therapeutic programs within both our proprietary development and partnered portfolios. Our primary focus is on our lead program MOR208, and here we look forward to releasing latest data from the ongoing L-MIND trial in an oral presentation at the forthcoming ASH (Free ASH Whitepaper) 2018 meeting. This program is proceeding according to plan, and we are committed to completing L-MIND and seeking U.S. approval based on this trial," commented Dr. Simon Moroney, CEO of MorphoSys AG. "Meanwhile, we continue to prepare our commercial organization in the U.S., with the goal of launching MOR208 there, subject, of course, to FDA approval of this investigational drug."

"We are very pleased with MorphoSys’s business performance in 2018 to date. Driven by an attractive licensing deal with Novartis for MOR106 and increasing royalty income from Tremfya(R), we saw strong revenue development in the third quarter. The very good business performance enabled us to increase our financial guidance for the year. Based on a solid cash position, which we further strengthened with our Nasdaq IPO earlier in the year, we are well positioned to continue the advancement of our pipeline products, in particular to drive our lead program MOR208 towards the market," said Jens Holstein, CFO of MorphoSys AG.

Financial review for the third quarter of 2018 (IFRS; all figures rounded)

In Q3 2018, MorphoSys continued to focus on the research and development of drug candidates in its proprietary portfolio, while also supporting the activities of its partners. Group revenues in Q3 2018 amounted to EUR 55.0 million (Q3 2017: EUR 15.0 million). The revenue increase was mainly driven by the up-front payment of EUR 47.5 million for the license agreement for MOR106 with Novartis. As the contractual royalty reporting from Janssen for Q3 2018 has not yet been received due to the reporting schedules of Janssen and MorphoSys, Tremfya(R) royalties booked for Q3 2018 were estimated based on public announcements made by Janssen/J&J on Tremfya(R) sales in Q3 2018.

In the Proprietary Development segment, MorphoSys focuses on the research and clinical development of its own drug candidates in the fields of cancer and inflammation. In Q3 2018, this segment recorded revenues of EUR 48.8 million (Q3 2017: EUR 0.2 million). In the Partnered Discovery segment, MorphoSys applies its proprietary technology to discover new antibodies for pharmaceutical companies, benefiting from its partners’ development advances through R&D funding, licensing fees, success-based milestone payments and royalties. In Q3 2018, revenues in this segment amounted to EUR 6.2 million (Q3 2017: EUR 14.8 million).

Total operating expenses reached EUR 25.3 million in the third quarter of 2018 (Q3 2017: EUR 38.2 million). R&D expenses for proprietary development and technology development amounted to EUR 15.9 million (Q3 2017: EUR 29.8 million).

Earnings before interest and taxes (EBIT) in Q3 2018 amounted to EUR 30.1 million (Q3 2017: EUR -23.5 million), reflecting in particular the up-front payment made by Novartis under the MOR106 license agreement. The Proprietary Development segment reported an EBIT of EUR 30.3 million (Q3 2017: EUR -29.8 million), while the Partnered Discovery segment recorded an EBIT of EUR 3.8 million (Q3 2017: EUR 10.4 million). In Q3 2018, the consolidated net result amounted to EUR 30.2 million (Q3 2017: EUR -24.0 million). Basic earnings per share for Q3 2018 reached EUR 0.96 (Q3 2017: EUR -0.83).

At the end of Q3 2018, the Company had a cash position of EUR 481.2 million, compared to EUR 312.2 million on December 31, 2017. On the balance sheet, this cash position is reported under the following items: cash and cash equivalents; financial assets at fair value through profit or loss; and current and non-current other financial assets at amortized cost. The increase in funds resulted mainly from the capital increase in conjunction with the successful Nasdaq listing completed in April 2018 with gross proceeds of USD 239 million and the upfront payment made by Novartis in the third quarter 2018 in the amount of EUR 47.5 million in connection with the license agreement for MOR106. This was partially offset by the use of cash for operating activities.

The number of shares issued totaled 31,839,572 at the end of Q3 2018 (year-end 2017: 29,420,785). The increase was mainly driven by the capital increase in April 2018.

Results for the first nine months 2018

For the first nine months of 2018, group revenues amounted to EUR 66.0 million (Q1-Q3 2017: EUR 38.6 million). Expenditure for proprietary development and technology development amounted to EUR 55.1 million in the first nine months of 2018 (Q1-Q3 2017: EUR 67.1 million). Consequently, EBIT in the first nine months of 2018 amounted to EUR -13.0 million, compared to EUR -53.8 million in the first nine months of 2017.

Financial guidance and operational outlook for 2018

MorphoSys confirms its 2018 financial guidance which had been increased after signing an agreement with Novartis for MOR106 in July 2018. In the light of the recent positive development of Tremfya(R) royalties, MorphoSys expects revenues on the upper end of the guided range from EUR 67 million to EUR 72 million for 2018. Earnings before interest and taxes (EBIT) are expected to be EUR -55 million to EUR -65 million. R&D expenses for proprietary programs and technology development are expected to be in a range of EUR 87 million to EUR 97 million. This guidance does not include additional revenues from potential future collaborations and/or license agreements nor any effects from possible in-licensing or development partnerships for new drug candidates.

MorphoSys expects the following events and activities in the Proprietary Development segment during the remainder of the year:

MOR208

– L-MIND:

– Continue discussions with the FDA to evaluate possible paths to market, including the possibility of an expedited regulatory submission and potential approval based primarily on the L-MIND study.

– Presentation of updated interim results on all 81 patients enrolled in the study evaluating MOR208 plus lenalidomide in r/r DLBCL at the ASH (Free ASH Whitepaper) (American Society of Hematology) 2018 Annual Meeting, which will be held in San Diego, California, in early December.

– B-MIND: Continue the enrollment in the phase 3 part of the study evaluating MOR208 plus bendamustine versus rituximab plus bendamustine in r/r DLBCL.

– COSMOS: Continue the phase 2 trial of MOR208 plus idelalisib or venetoclax in chronic lymphocytic leukemia (CLL/SLL) and present data from cohort B (MOR208 plus venetoclax) at the upcoming ASH (Free ASH Whitepaper) 2018 Annual Meeting.

– Commercial and CMC activities: Secure commercial supply of MOR208 and continue to build commercial capabilities for MOR208 in the U.S. under the newly established MorphoSys US Inc., in preparation for a potential market launch, currently anticipated in 2020 pending FDA approval.

MOR202

– Multiple myeloma (MM): Study results from the ongoing phase 1/2a study will also be presented at the upcoming ASH (Free ASH Whitepaper) 2018 Annual Meeting. As previously communicated, MorphoSys will not continue development in MM beyond completion of the current trial without an additional partner for Ex-China; MorphoSys expects its partner I-Mab Biopharma to continue preparations for clinical development in MM and to start a pivotal trial in early 2019 in China.

– Other indications: MorphoSys continues to evaluate the development of MOR202 in other indications including autoimmune disorders.

MOR106: Continue ongoing development activities with partner Galapagos under the new global licensing agreement with Novartis.

– Continue the ongoing phase 2 trial IGUANA in atopic dermatitis.

– Continue the phase 1 bridging study initiated in September 2018 to evaluate a subcutaneous formulation of MOR106.

– All future costs related to MOR106 development to be borne by Novartis.

MOR107: Continue preclinical investigations of MOR107 with a focus on oncology indications.

MOR103/GSK3196165: Following presentation of data from several phase 2 trials in inflammatory diseases including rheumatoid arthritis (RA) run by partner GlaxoSmithKline (GSK) at the American College of Rheumatology (ACR) meeting in October 2018, MorphoSys expects GSK to further continue clinical development activities in RA.

In its Partnered Discovery segment, MorphoSys expects the following events in the fourth quarter of 2018:

Tremfya(R) (guselkumab): Several phase 3 trials in psoriasis are scheduled for primary completion in 2018 according to clinicaltrials.gov. This includes a head-to-head trial comparing Tremfya(R) to Cosentyx(R) in adults with plaque psoriasis, results of which are expected to be communicated in early 2019. MorphoSys expects Janssen to continue its current development program with Tremfya(R) including two phase 3 trials in psoriatic arthritis, the phase 2/3 GALAXI program in Crohn’s disease as well as the clinical phase 3 PROTOSTAR trial in pediatric psoriasis patients.

Other partnered programs: The publication of clinical data and the achievement of regulatory milestones from other partnered programs may occur during the remainder of 2018.

MorphoSys will continue to support its proprietary development activities by evaluating potential in-licensing, co-development, and/or acquisition opportunities as well as by initiating new proprietary development programs with the goal of maintaining and expanding the Company’s position in its current therapeutic and technological fields of activities.

** Including MOR107, which concluded a phase 1 study in 2017 and is currently in preclinical investigation with a focus on oncology indications. Tremfya(R) is still considered as a clinical program due to ongoing studies in various indications.
*** Including MOR103/GSK3196165, which is fully out-licensed to GSK, and MOR106, for which MorphoSys and Galapagos have signed a global licensing agreement with Novartis.

The interim statement for the third quarter of 2018 (IFRS) is available online:

View Source

MorphoSys will hold a conference call and webcast on November 6, 2018, to present the third quarter 2018 financial results and provide an outlook for the remainder of 2018.

Dial-in number for the analyst conference call (in English) at 2:00pm CET; 1:00pm GMT; 8:00am EST (listen-only):

Germany: +49 (0) 69 201 744 210

For UK residents: +44 (0) 203 009 2470

For US residents: +1 (0) 877 423 0830

Participant PIN: 79499880#

Participants are asked to dial in 10 minutes before the beginning of the conference. A live webcast and slides will be made available at View Source After the conference call, a slide-synchronized audio replay of the conference and a transcript will be available at View Source

Ultragenyx Reports Third Quarter 2018 Financial Results and Corporate Update

On November 5, 2018 Ultragenyx Pharmaceutical Inc. (NASDAQ: RARE), a biopharmaceutical company focused on the development of novel products for serious rare and ultra-rare genetic diseases, reported its financial results and corporate update for the quarter ended September 30, 2018 (Press release, Ultragenyx Pharmaceutical, NOV 5, 2018, View Source [SID1234530762]).

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’re encouraged by the growing demand for Crysvita from both children and adults with XLH in the United States, and we are making significant progress reaching doctors and helping them get patients on commercially available therapy in this early launch phase," said Emil D. Kakkis, M.D., Ph.D., Chief Executive Officer and President of Ultragenyx. "Next year, we expect to file for potential approval of our third therapy, UX007 in fatty acid oxidation disorders, and we continue to advance our two gene therapy clinical programs with additional data expected around the end of this year and in 2019."

Financial Results

For the third quarter of 2018, Ultragenyx reported a net loss of $87.3 million, or $1.74 per share, basic and diluted, compared with a net loss for the third quarter of 2017 of $79.2 million, or $1.87 per share, basic and diluted. For the nine months ended September 30, 2018, net loss was $109.8 million, or $2.22 per share, basic and diluted, compared with a net loss for the same period in 2017 of $220.4 million, or $5.22 per share, basic and diluted. The loss from the first nine months was reduced by the sale of the Mepsevii (vestronidase alfa) priority review voucher (PRV) in January 2018 for net proceeds of $130.0 million and a $40.3 million gain from Ultragenyx’s portion of the sales of the PRV received with the Crysvita (burosumab) approval. The net loss for the first nine months of 2018 reflected cash used in operations of $234.7 million compared to $172.0 million for the same period in 2017.

Net Revenues

For the third quarter of 2018, Ultragenyx reported $11.8 million in total revenue. For Crysvita, Ultragenyx recognized $5.4 million in profit sharing and royalty revenue from its collaboration and license agreement with Kyowa Hakko Kirin. This includes $4.4 million in collaboration revenue in the U.S. profit share territory and $1.0 million in royalty revenue in the European territory. Net product sales for Crysvita in other regions were nominal. Mepsevii product revenue for the third quarter of 2018 was $2.1 million, and UX007 named patient revenue was $0.4 million. Ultragenyx recognized $3.6 million in revenue from its research agreement with Bayer.

Operating Expenses

Total operating expenses for the third quarter of 2018 were $101.4 million compared with $83.9 million for the same period in 2017, including non-cash stock-based compensation of $20.7 million and $17.2 million in the third quarter of 2018 and 2017, respectively. Total operating expenses for the nine months ended September 30, 2018, were $316.3 million compared with $232.3 million for the same period in 2017, including non-cash stock-based compensation of $59.0 million and $48.5 million in the first nine months of 2018 and 2017, respectively. The increase in total operating expenses is due to the increase in commercial, development, and general and administrative costs as the company commercializes, grows and advances its pipeline.

Cash, Cash Equivalents and Investments

Cash, cash equivalents and investments were $503.1 million as of September 30, 2018.

Recent Updates

Crysvita in X-Linked Hypophosphatemia (XLH)

Ultragenyx submitted regulatory filings in Canada, Brazil and Colombia. We anticipate regulatory decisions in these markets over the course of 2019. Reimbursed named patient treatment has already begun in Argentina in response to physician requests for multiple patients.
Crysvita in Tumor-Induced Osteomalacia (TIO)

Positive longer-term 48-week and 72-week data from the Phase 2 study of Crysvita in adults with TIO were presented at the American Society for Bone and Mineral Research (ASBMR) 2018 Annual Meeting in Montreal. In adults with TIO, Crysvita was associated with increases in serum phosphorus and serum 1,25 dihydroxy vitamin D levels; improvements in osteomalacia, mobility and vitality; and reductions in fatigue. Adverse events (AE) generally reflected the patients’ underlying diseases, and there were no serious treatment-related AEs during the study. Discussions with the U.S. Food and Drug Administration (FDA) regarding the regulatory pathway are ongoing.
Mepsevii in Mucopolysaccharidosis VII (MPS VII)

The European Commission (EC) approved the Marketing Authorization Application (MAA) for Mepsevii under exceptional circumstances for the treatment of non-neurological manifestations of MPS VII. Mepsevii is now approved for use in all 28 EU countries as well as in Iceland, Liechtenstein and Norway, and has launched in Germany.
Brazil’s National Health Surveillance Agency approved Mepsevii for the treatment of MPS VII for patients of all ages. Additional regulatory decisions for patients in Columbia and Chile are anticipated over the course of 2019.
UX007 in Long-Chain Fatty Acid Oxidation Disorders (LC-FAOD) and Glucose Transporter Type 1 Deficiency Syndrome (Glut1 DS)

The FDA accepted Ultragenyx’s proposal to submit a New Drug Application (NDA) for UX007 for the treatment of LC-FAOD based on currently available data. Further details will be forthcoming following a pre-NDA meeting, which is scheduled to take place by the end of 2018. Additionally, discussions are progressing with the European Medicines Agency regarding a potential conditional marketing authorization in Europe.

In October 2018, Ultragenyx announced that the Phase 3 study evaluating UX007 in patients with Glut1 DS did not achieve its primary endpoint compared to placebo. The safety profile observed in this study was consistent with what has been previously reported with UX007. Ultragenyx plans to discontinue further Glut1 DS clinical development for UX007, and expects no impact on plans for the LC-FAOD indication.
DTX301 Gene Therapy in Ornithine Transcarbamylase (OTC) Deficiency

Positive topline data from the first two dose cohorts of the Phase 1/2 study demonstrate normalization of ureagenesis in two patients and further support proof-of-concept. The first responder (Cohort 1) showed a further increased level of ureagenesis at 52 weeks and has been clinically stable for more than eight months after discontinuing alternate pathway medications and also after liberalizing a protein-restricted diet near the 52 week time point. The second responder (Cohort 2) achieved normalization of ureagenesis at 24 weeks, and has been clinically stable for more than one month after also discontinuing all alternate pathway medications. The remaining four patients in Cohorts 1 and 2 continue to demonstrate no clinically meaningful change in rate of ureagenesis. All patients have remained clinically and metabolically stable, with only modest rises in ALT well treated by a reactive, tapering steroid regimen. The first patient in cohort 3 has been dosed. Data from higher dose Cohort 3 are expected in mid-2019.
DTX401 Gene Therapy in Glycogen Storage Disease Type Ia (GSDIa)

All three patients in the lowest-dose Cohort 1 of the Phase 1/2 study have been dosed. Data from this cohort are expected around the end of 2018.
CDKL5 Deficiency Disorder (CDD)

Ultragenyx exercised its option with REGENXBIO Inc. to develop a gene therapy to treat patients with CDD using REGENXBIO’s adeno-associated virus (AAV) vectors, including AAV9. CDD is a severe and debilitating neurological disorder that shares many features of Rett Syndrome. It was commonly identified as an atypical Rett Syndrome until improved genetic testing led to more accurate diagnosis and an increasing prevalence of diagnosed CDD-affected patients.
Corporate

Ultragenyx appointed a Chief Operating Officer, Wladimir (Vlad) Hogenhuis, M.D., on September 28, 2018. In this newly created role, Dr. Hogenhuis oversees the Global Commercial, Business Development, and Manufacturing organizations.
Conference Call and Webcast Information

Ultragenyx will host a conference call today, Monday, November 5, 2018, at 2 p.m. PT/ 5 p.m. ET to discuss third quarter 2018 financial results and provide a corporate update. The live and replayed webcast of the call will be available through the company’s website at View Source To participate in the live call by phone, dial (855) 797-6910 (USA) or (262) 912-6260 (international) and enter the passcode 1290627. The replay of the call will be available for one year.

BioCryst to Present Abstracts at Annual Scientific Meeting of American College of Allergy, Asthma & Immunology

On November 5, 2018 BioCryst Pharmaceuticals, Inc. (Nasdaq:BCRX) reported that the company will present three abstracts at the upcoming Annual Scientific Meeting of the American College of Allergy, Asthma & Immunology (ACAAI) November 15-19 in Seattle (Press release, BioCryst Pharmaceuticals, NOV 5, 2018, View Source [SID1234530749]).

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!

Oral Administration of a Liquid Formulation of BCX7353 Rapidly Provides Sustained Concentrations and Kallikrein Inhibition; Poster P153, Friday, November 16, 3:30 p.m. PT

Relationship of Target Concentrations with Efficacy: Results from the APeX-1 Study of BCX7353; Poster P157, Friday, November 16, 4:10 p.m. PT

Pharmacokinetics and Pharmacodynamics of BCX7353, an Oral Plasma Kallikrein Inhibitor, in Healthy Japanese Subjects, Poster P158, Friday, November 16, 4:20 p.m. PT
"The Annual Scientific Meeting of the ACAAI provides a focused opportunity for us to meet with many of the clinical and academic leaders driving continued innovation to improve HAE therapy, and we are excited to, once again, have an important scientific presence at the meeting," said Dr. William Sheridan, chief medical officer of BioCryst.

At the 60th Annual Meeting of the American Society of Hematology, Takeda will present results on a broad spectrum of blood cancer therapy needs

On November 5, 2018 Takeda Pharmaceutical Company Limited ( TSE: 4502 ) reported that 18 corporate-sponsored abstracts will be presented at the 60th Annual Meeting of the American Society of Hematology (ASH) (Free ASH Whitepaper) , which will be held December 1-20, 2018 in San Diego (Press release, Takeda, NOV 5, 2018, View Source [SID1234530741]). Takeda’s presentations will present new clinical trial data from the company’s hematology portfolio. In particular, Takeda reports results from the Phase 3 clinical trials TOURMALINE-MM3 and ECHELON-2.

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!

"By providing data from two Phase 3 clinical trials and from the Company’s pipeline, Takeda continues to expand the evidence base for new therapeutic options that improve the treatment of blood cancer patients," Dr. Takeda said. Christophe Bianchi, MD, President, Takeda Global Oncology Business Unit. "Positive results from the TOURMALINE-MM3 trial, the first and only placebo-controlled phase 3 trial to test a proteasome inhibitor in this setting, showed that NINLARO maintenance therapy after autologous stem cell transplantation improved progression-free survival compared to the control arm. Thus, NINLARO can potentially be used as a maintenance therapy in a patient group, currently limited treatment options are available. In addition, the positive results of the ECHELON-2 study demonstrated that ADCETRIS in combination with chemotherapy provided better progression-free survival and overall survival in patients with previously untreated CD30-positive peripheral T-cell lymphoma than the control arm. This is an important milestone for ADCETRIS as one possible treatment option in this setting, where standard care has been unchanged for several decades. " that ADCETRIS in combination with chemotherapy achieved better progression-free survival and overall survival in patients with previously untreated CD30-positive peripheral T-cell lymphoma than the control arm. This is an important milestone for ADCETRIS as one possible treatment option in this setting, where standard care has been unchanged for several decades. " that ADCETRIS in combination with chemotherapy achieved better progression-free survival and overall survival in patients with previously untreated CD30-positive peripheral T-cell lymphoma than the control arm. This is an important milestone for ADCETRIS as one possible treatment option in this setting, where standard care has been unchanged for several decades. "

At this year’s ASH (Free ASH Whitepaper) meeting, results of the Phase 3 TOURMALINE-MM3 study addressing the effect of NINLARO (ixazomib) maintenance therapy in adult patients with multiple myeloma responding to high-dose therapy (HDT) and autologous stem cell transplantation (ASCT), examined, presented for the first time in a lecture. The lecture will be held on Sunday, December 2nd at 7:30 pm PT. The TOURMALINE MM3 trial achieved its primary endpoint with NINLARO, which led to a statistically significant improvement in progression-free survival (PFS) compared to placebo, assessed by an Independent Review Committee (IRC). There were no new safety signals in the TOURMALINE MM3 study and the safety profile of NINLARO maintenance therapy is in line with the already published results on the use of NINLARO as monotherapy. NINLARO is not currently approved as a single agent for ASCT maintenance therapy.

Results of the Phase 3 ECHELON-2 study will be presented in a lecture on Monday, December 3 at 6:15 pm PT. The study found a statistically significant improvement in PFS for ADCETRIS (brentuximab vedotin) in combination with CHP (cyclophosphamide, doxorubicin, prednisone) compared to the control arm, CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone). Main results of the ECHELON-2 study were reported in October 2018. Study results showed that combination therapy with ADCETRIS plus CHP for progression-free survival was superior to the control arm as assessed by an Independent Review Facility (IRF) (hazard ratio = 0.71, p = 0.0110). For all important secondary endpoints, including overall survival, there were statistically significantly better outcomes for the ADCETRIS-plus-CHP arm with a manageable safety profile. ADCETRIS is currently not approved as a PTCL frontline therapy.

The breadth and depth of Takeda’s research and development efforts will also be highlighted in several presentations focusing on multiple myeloma, lymphoma, chronic myeloid leukemia, and myelodysplastic syndromes (MDS).

Eighteen abstracts sponsored by Takeda were accepted for presentation at the ASH (Free ASH Whitepaper) Annual Meeting in 2018, including:

Note: All times are in Pacific Standard Time (PT).

ADCETRIS (Brentuximab Vedotin)

The ECHELON-2 study: Results of a randomized, double-blind, drug-controlled Phase 3 study of brentuximab vedotin and CHP (A + CHP) versus CHOP in the frontline treatment of patients with CD30-positive peripheral T-cell lymphoma . Abstract 997. Lecture. Monday, December 3, 2018, 6:15 – 7:45 pm (San Diego Convention Center, Room 6F).
Elderly patients with previously untreated classic Hodgkin’s lymphoma (cHL): An in-depth analysis of the Phase 3 ECHELON-1 study . Abstract 1618. Saturday, December 1, 2018, 6:15 – 8:15 pm (San Diego Convention Center, Hall GH).
Superior clinical benefit of brentuximab vedotin in mycosis fungoides versus physician’s choice, regardless of CD30 grade or large cell transformation status in the Phase 3 ALCANZA study . Abstract 1646. Saturday, December 1, 2018, 6:15 pm – 8:15 pm (San Diego Convention Center, Hall GH).
Brentuximab Vedotin with chemotherapy in adolescents and young adults with stage III or IV Hodgkin’s lymphoma: A subgroup analysis of the Phase 3 ECHELON-1 study . Abstract 1647. Saturday, December 1, 2018, 6:15 – 8:15 pm (San Diego Convention Center, Hall GH).
Brentuximab Vedotin plus chemotherapy in patients with advanced classical Hodgkin’s lymphoma (cHL): testing of modified progression-free survival (mPFS) and conventional PFS in the Phase 3 ECHELON-1 study . Abstract 2904. Sunday, December 2, 2018, 6:00 pm – 8:00 pm (San Diego Convention Center, Hall GH).
Regression of peripheral neuropathies (PN) in patients receiving A + AVD or ABVD in the Phase 3 ECHELON-1 study . Abstract 2921. Sunday, December 2, 2018, 6:00 pm – 8:00 pm (San Diego Convention Center, Hall GH).
Phase 1 results of a phase 1/2 study evaluating the safety, tolerability and recommended Phase 2 dose (RP2D) of brentuximab vedotin plus doxorubicin, vinblastine and dacarbazine (A + AVD) in pediatric patients with advanced, new diagnosed classical Hodgkin’s lymphoma (cHL) . Abstract 1644. Saturday, December 1, 2018, 6:15 – 8:15 pm (San Diego Convention Center, Hall GH).
Multiple Myeloma / NINLARO (Ixazomib)

Maintenance therapy with oral proteasome inhibitor (PI) Ixazomib causes significant progression-free survival (PFS) after autologous stem cell transplantation (ASC) in patients with newly diagnosed multiple myeloma (NDMM): Phase 3 TOURMALINE-MM3 study . Abstract 301. Lecture. Sunday, December 2, 2018, 7:30 am – 9:00 am (Marriott Marquis San Diego Marina, Grand Ballroom 7)
Addition of ixazomib to an Rd backbone improves clinical benefit in patients with relapsed / refractory multiple myeloma (RRMM) with non-canonical NF-κB activation – results of the TOURMALINE MM1 study . Abstract 473. Lecture. Sunday, December 2, 2018, 4:30 – 6:00 pm (Marriott Marquis San Diego Marina, Grand Ballroom 7).
Treatment preferences of patients with relapsed / refractory multiple myeloma: Are patients ready for a compromise between efficacy and tolerability? Abstract 614th Lecture. Monday, December 3, 2018, 7:00 am – 8:30 am (San Diego Convention Center, Room 11B).
Ixazomib plus lenalidomide-dexamethasone (IRd) in patients with relapsed / refractory multiple myeloma (MM) – day-to-day efficacy is similar to efficacy in the Phase 3 TOURMALINE-MM1: a pooled analysis of the INSIGHT MM and the Czech Register Monoclonal gammopathies (RMG) . Abstract 1971. Saturday, December 1, 2018, 6:15 – 8:15 pm (San Diego Convention Center, Hall GH).
Socio-demographic characteristics and societal perspective in patients with relapsed and / or refractory multiple myeloma (RRMM) in Spain: an interim analysis of the CHARISMMA study . Abstract 2300. Saturday, December 1, 2018, 6:15 pm – 8:15 pm (San Diego Convention Center, Hall GH).
Transplant status without influence on the choice of induction schemes in patients with newly diagnosed multiple myeloma (NDMM) in the prospective observational study INSIGHT MM . Abstract 3289. Sunday, December 2, 2018, 6:00 pm – 8:00 pm (San Diego Convention Center, Hall GH).
Dynamic changes in the International Staging System as a predictor of survival outcome in patients with advanced multiple myeloma . Abstract 4438. Monday, December 3, 2018, 6:00 pm – 8:00 pm (San Diego Convention Center, Hall GH).
ICLUSIG (Ponatinib)

Comparing cardiovascular events in various tyrosine kinase inhibitors in patients with chronic myeloid leukemia from clinical practice . Abstract 3567. Sunday, December 2, 2018, 6:00 pm – 8:00 pm (San Diego Convention Center, Hall GH).
Pipeline (lymphoma, multiple myeloma, myelodysplastic syndromes)

Patient characteristics and treatment patterns in first- and second-line therapy in diffuse large B-cell lymphoma and follicular lymphoma in the United Kingdom, France, and Germany . Abstract 4234. Monday, December 3, 2018, 6:00 pm – 8:00 pm (San Diego Convention Center, Hall GH).
A single dose of the cytolytic CD38 antibody TAK-079 in healthy volunteers: tolerability, pharmacokinetics, and pharmacodynamics . Abstract 3249. Sunday, December 2, 2018, 6:00 pm – 8:00 pm (San Diego Convention Center, Hall GH).
Review patient-reported outcomes in patients with myelodysplastic syndromes: Can a tailor-made selection of EORTC library items improve the EORTC QLQ-C30? Abstract 4856. Monday, December 3, 2018, 6:00 pm – 8:00 pm (San Diego Convention Center, Hall GH).
For more information, the ASH (Free ASH Whitepaper) program is available here: View Source

About ADCETRIS
ADCETRIS is an ADC derived from a monoclonal anti-CD30 antibody that uses proprietary technology from Seattle Genetics to bind a protease cleavable linker to monomethylauristatin E (MMAE), a microtubule-disrupting agent. The linker system of the ADC is designed to remain stable in the bloodstream. Only after internalization of the conjugate into the CD30-positive tumor cells MMAE is released.

The ADCETRIS intravenous infusion injection has received FDA approval for five indications for the treatment of adult patients: (1) untreated classical Hodgkin lymphoma (cHL) stage III or IV, in combination with chemotherapy, (2) high cHL Recurrence or progression risk as a consolidation after autologous hematopoietic stem cell transplantation (auto-HSCT); (3) cHL after auto HSCT failure or failure of at least two prior polychemotherapy regimens in patients not eligible for auto HSCT;(4) sALCL after failure of at least one prior polychemotherapy and (5) primary cutaneous anaplastic large cell lymphoma (pcALCL) or CD30-expressing mycosis fungoides (MF) in patients who have previously received systemic therapy.

Health Canada approved ADCETRIS for relapsed or refractory Hodgkin’s lymphoma and sALCL in 2013, and granted ADCETRIS full approval for autologous stem cell transplantation (ASCT) consolidation therapy in patients with Hodgkin’s lymphoma at increased risk of recurrence or progression.

ADCETRIS received conditional marketing approval from the European Commission in October 2012. The indications approved in Europe are: (1) for the treatment of adult patients with relapsed or refractory CD30-positive Hodgkin’s lymphoma after ASCT or after at least two previous therapies, when ASCT or polychemotherapy are not treatment options, (2) for the treatment of adult patients with relapsed or refractory sALCL, (3) for the treatment of adult patients with CD30-positive Hodgkin’s lymphoma who are at increased risk of recurrence or progression after ASCT, and (4) for the treatment of adult patients with CD30-positive cutaneous T-cell Lymphoma (CTCL) after at least one previous systemic therapy.

ADCETRIS has been approved by the regulatory authorities in more than 70 countries for relapsed or refractory Hodgkin’s lymphoma and sALCL. Please observe the following important safety instructions.

ADCETRIS is currently being extensively studied in more than 70 ongoing clinical trials, including a Phase 3 study for first-line treatment in Hodgkin’s lymphoma (ECHELON-1) and another Phase 3 trial for use as a frontline therapy CD30-positive peripheral T-cell lymphoma (ECHELON-2) as well as studies on numerous other types of CD30-positive malignancies.

Seattle Genetics and Takeda jointly develop ADCETRIS. Under the terms of the cooperation agreement, Seattle Genetics holds the marketing rights to ADCETRIS in the US and Canada, while Takeda owns the marketing rights in the rest of the world. Seattle Genetics and Takeda are sharing the co-development costs of ADCETRIS in equal parts, with the sole exception of Japan being responsible for development costs.

Important Safety Information of ADCETRIS (Brentuximab Vedotin) (European Union)

Please refer to the Specialist Information (SmPC) before a prescription.

CONTRAINDICATIONS

ADCETRIS is contraindicated in patients with hypersensitivity to brentuximab vedotin and its other ingredients. In addition, the concomitant use of ADCETRIS and bleomycin causes pulmonary toxicity.

SPECIAL WARNINGS AND PRECAUTIONS

Progressive multifocal leukoencephalopathy (PML): In ADCETRIS-treated patients, reactivation of the John Cunningham virus (JCV) may lead to progressive multifocal leukoencephalopathy (PML) and subsequent death. PML has been reported in patients who received ADCETRIS after receiving several other chemotherapy regimens. PML is a rare demyelinating disease of the central nervous system, which is caused by a reactivation of a latent JCV and often leads to death.

Patients should be closely monitored for new or worsening neurological, cognitive, or behavioral signs or symptoms that may indicate PML. PML suspicion options include neurological examination, brain gadolinium contrast MRI, and CSF DNA CSF analysis by polymerase chain reaction or brain biopsy with detection of JCV. A negative JCV PCR does not rule out PML. If no alternative diagnosis can be made, further follow-up and clarification may be indicated. The administration of ADCETRIS should be suspended whenever PML is suspected, and ADCETRIS must be discontinued if the PML diagnosis is confirmed.

Attention should be paid to symptoms of PML that may not be noticed by the patient (eg cognitive, neurological or psychiatric symptoms).

Pancreatitis: Acute pancreatitis has been reported in ADCETRIS-treated patients, sometimes with fatal outcome. Patients should be monitored closely for newly occurring or worsening abdominal pain, which may indicate acute pancreatitis. Patient examinations should include physical examinations, laboratory tests for serum amylase and serum lipase, and abdominal imaging such as ultrasound and other appropriate diagnostic methods. If acute pancreatitis is suspected, ADCETRIS should be discontinued. If the diagnosis of acute pancreatitis is confirmed, ADCETRIS should be discontinued permanently.

Pulmonary toxicity: Cases of pulmonary toxicity such as pneumonitis, interstitial lung disease and acute respiratory distress syndrome (ARDS) have been reported in patients receiving ADCETRIS, some of which are fatal. Although a causal relationship with ADCETRIS is not established, the risk of pulmonary toxicity can not be excluded. New or worsening pulmonary symptoms should be clarified immediately and treated accordingly. During the clarification and until the improvement of the symptoms, an interruption of the administration should be considered.

Serious infections and opportunistic infections: ADCETRIS-treated patients have experienced severe infections such as pneumonia, staphylococcal bacteremia, sepsis or septic shock (including fatal outcomes) and herpes zoster, and opportunistic infections such as Pneumocystis pneumonia ( Pneumocystis jiroveci ) and oral candidosis. Patients should be monitored carefully for possible signs of severe or opportunistic infection during treatment.

Infusion-Related Reactions (IRR): In the ADCETRIS therapy, immediate and delayed infusion reactions as well as anaphylactic reactions have occurred. Patients should be monitored carefully during and after an infusion. If an anaphylactic reaction occurs, the administration of ADCETRIS should be stopped immediately and for good medical treatment. In the case of an IRR, the infusion should be discontinued and appropriate medical measures should be taken. The infusion may be restarted at a slower rate once symptoms have resolved. Patients already experiencing an infusion-related reaction should be appropriately premedicated for subsequent infusions.

Tumor lysis syndrome (TLS): Cases of TLS have been reported in ADCETRIS. Patients with rapidly proliferating tumors and high tumor burden are at risk for TLS. These patients should be closely monitored and treated with the most appropriate medical procedures.

Peripheral neuropathy (PN): Treatment with ADCETRIS may cause sensory or motor PN. ADCETRIS-induced peripheral neuropathy is typically cumulative and, in most cases, reversible. Patients should be monitored for signs of PN, such as hypoaesthesia, hyperesthesia, paraesthesia, malaise, burning sensation, neuropathic pain or weakness. In patients who experience a new or worsening PN, the dose may need to be delayed and reduced or ADCETRIS discontinued.

Hematologic Toxicities : ADCETRIS may have Grade 3 or 4 anemia, thrombocytopenia, and persistent (or more than one week) neutropenia of Grade 3 or 4. Prior to administration of each dose, the large blood count should be monitored.

Febrile Neutropenia: Cases of febrile neutropenia have been reported. Patients should be closely monitored for fever and treated with the most appropriate medical procedures if febrile neutropenia develops.

Stevens-Johnson syndrome (SJS): Cases of SJS and toxic epidermal necrolysis (TEN) have been reported in ADCETRIS therapy, some of them fatal. If an SJS or TEN occurs, treatment with ADCETRIS must be discontinued and appropriate medical treatment initiated.

Gastrointestinal complications: Gastrointestinal complications, sometimes fatal, have been reported, including intestinal obstruction, ileus, enterocolitis, neutropenic colitis, erosions, ulcers, perforations and bleeding. New or worsening gastrointestinal complaints should be clarified immediately and treated accordingly.

Hepatotoxicity: Increased alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels have been reported, including severe cases of liver toxicity, some of which are fatal. Liver function should be monitored in patients receiving ADCETRIS before initiating therapy and then periodically for elevated liver function. In patients with hepatotoxicity, a delay or change in dose or discontinuation of ADCETRIS therapy may be required.

Hyperglycaemia: There have been cases of hyperglycaemia in patients with an increased body mass index (BMI) with or without a history of diabetes mellitus. In patients experiencing a hyperglycaemic event, serum glucose levels should be monitored closely. If necessary, appropriate antidiabetic treatment should be used.

Renal and hepatic impairment: There is limited experience in patients with impaired renal and hepatic function. Available data suggest that MMAE excretion could be compromised by severe renal or hepatic dysfunction and low serum albumin concentrations.

CD30-positive CTCL: The extent of treatment effect on CD30-positive CTCL subtypes other than mycosis fungoides (MF) and primary cutaneous anaplastic large cell lymphoma (pcALCL) is not clear due to a lack of evidence at high evidence levels. Disease activity was demonstrated in two single-arm Phase 2 studies of ADCETRIS in Sézary syndrome (SS) subtypes, lymphomatoid papulosis (LyP), and mixed CTCL histology. These data suggest that efficacy and safety can be extrapolated for other CD30-positive CTCL subtypes. A careful risk-benefit assessment should be made for each patient, and caution should be exercised in other types of CD30-positive CTCL patients.

Sodium content of excipients : ADCETRIS contains a maximum of 2.1 mmol (47 mg) sodium per dose. This should be considered in patients with controlled sodium diet.

INTERACTIONS
Patients receiving strong CYP3A4 and P-gp inhibitors concomitantly with ADCETRIS may be at increased risk of neutropen- sation, so these patients should be closely monitored. The concomitant administration of ADCETRIS and a CYP3A4 inducer did not alter the plasma exposure of ADCETRIS, but appeared to reduce the plasma concentration of MMAE metabolites that could be analyzed. It is not expected that ADCETRIS will affect exposure to medicinal products metabolised by CYP3A4 enzymes.

PREGNANCY: Women of childbearing potential are advised to use two reliable birth control methods during and up to six months after treatment with ADCETRIS. So far, there are no data on the use of ADCETRIS in pregnant women. However, animal studies have shown reproductive toxicity. ADCETRIS should not be used during pregnancy unless the potential benefit to the mother is significantly greater than the potential risk to the unborn child.

TERM: There is no data on whether ADCETRIS or its metabolites are excreted in breast milk. Therefore, a risk for the newborn / infant can not be excluded. Given the potential risk, a decision should be made as to whether weaning or discontinuation / abandonment of ADCETRIS therapy is more advisable.

FERTILITY: In non-clinical studies, treatment with ADCETRIS has been found to cause testicular toxicity and may therefore affect male fertility. Men treated with ADCETRIS are advised not to father a child during treatment and up to six months after the last dose.

Effects on ability to drive and use machines: ADCETRIS may have a minor influence on the ability to drive and use machines.

UNWANTED REACTIONS
The most common adverse reactions (> 10%) were infection, peripheral sensory neuropathy, nausea, fatigue, diarrhea, fever, upper respiratory tract infection, neutropenia, rash, cough, vomiting, arthralgia, peripheral motor neuropathy, infusion-related reactions, pruritus, Constipation, dyspnea, weight loss, myalgia and abdominal pain.

Serious adverse reactions included pneumonia, acute respiratory distress syndrome, headache, neutropenia, thrombocytopenia, constipation, diarrhea, vomiting, nausea, fever, motor and sensory peripheral neuropathy, hyperglycemia, demyelinating polyneuropathy, tumor lysis syndrome and Stevens-Johnson syndrome. Severe adverse reactions occurred in 12 percent of patients. The frequency of unique severe adverse reactions was ≤1 percent.

Important Safety Information for ADCETRIS (Brentuximab Vedotin) (USA)

FRAGRANTED WARNING: PROGRESSIVE MULTIFOCALE LEUKENCE PEPHALOPATHY (PML)

Patients receiving ADCETRIS may experience infection with the JC virus, resulting in PML and, subsequently, death.

Contraindication
Co-administration of ADCETRIS and bleomycin is contraindicated due to pulmonary toxicity (eg, interstitial infiltration and / or inflammation).

Warnings and Precautions

Peripheral Neuropathy (PN): Treatment with ADCETRIS causes a predominantly sensory PN. Cases of motor PN have also been reported. A PN triggered by ADCETRIS is cumulative. Patients should be monitored for the occurrence of symptoms such as hypoaesthesia, hyperesthesia, paraesthesia, discomforting and burning sensations, neuropathic pain or weakness. If these symptoms occur, appropriate dose changes should be made.

Anaphylactic and infusion-related reactions: Infusion reactions (IRR), including anaphylactic reactions, have occurred in association with ADCETRIS. Patients should be monitored during the infusion. In the case of an IRR, the infusion should be discontinued and appropriate medical measures should be taken. In an anaphylactic reaction, the infusion should be discontinued immediately and a suitable medical treatment should be performed. Patients who previously had an infusion reaction should be prepared for subsequent infusions. Premedication may include acetaminophen, an antihistamine and a corticosteroid.

Hematologic Toxicities : Under ADCETRIS, persistent (≥ 1 week) severe neutropenia and thrombocytopenia or Grade 3 or 4 anemia may occur. Cases of febrile neutropenia with ADCETRIS have been reported. Before each dose of ADCETRIS, a large blood count should be taken. Patients with Grade 3 or 4 neutropenia may be more likely to monitor more frequently. Patients should be monitored for fever. If Grade 3 or 4 neutropenia develops, delay or decrease in dose, discontinuation of therapy, or G-CSF prophylaxis should be considered at later doses.

Severe infections and opportunistic infections: Patients receiving ADCETRIS have been reported to have infections such as pneumonia, bacteremia and sepsis, or septic shock (including fatal cases). Patients should be monitored closely for possible bacterial, fungal or viral infections during treatment.

Tumor lysis syndrome: Patients with rapidly proliferating tumors and high tumor burden should be monitored closely.

Increased toxicity in severe renal insufficiency: Adverse reactions of grade 3 or higher and deaths were more common in patients with severe renal insufficiency than in patients with normal renal function. The use of ADCETRIS should be avoided in patients with severe renal insufficiency.

Increased toxicity in moderate or severe hepatic insufficiency: Grade 3 or higher adverse reactions and deaths were more common in patients with moderate or severe hepatic insufficiency than in patients with normal liver function. The use of ADCETRIS should be avoided in patients with moderate or severe hepatic insufficiency .

Hepatotoxicity : Patients receiving ADCETRIS have experienced severe cases of hepatotoxicity, some of which are fatal. The cases were consistent with hepatocellular damage, including elevated transaminase and / or bilirubin levels, and occurred after the first dose of ADCETRIS or re-exposure. Already existing liver disease, elevated baseline liver enzymes and concomitant medications may also increase the risk. Liver enzyme and bilirubin levels should be monitored. Patients with new, worsening or recurrent hepatotoxicity may require a delay, change in dose or discontinuation of ADCETRIS therapy.

Progressive multifocal leukoencephalopathy (PML): In ADCETRIS-treated patients, infections with the JC virus have been reported, leading to PML and subsequently death. The first symptoms occurred at various times after initiation of ADCETRIS therapy, with some occurring within three months of the first exposure. Other contributing factors besides ADCETRIS therapy include previous therapies as well as underlying diseases that could cause immunosuppression. The diagnosis of PML should be considered in all patients with emerging signs and symptoms of central nervous system disorders.

Pulmonary toxicity: Events of non-infectious pulmonary toxicity such as pneumonitis, interstitial lung disease and acute respiratory distress syndrome, among them fatal, have been reported. Patients should be monitored for signs and symptoms of pulmonary toxicity, including coughing and dyspnoea. In the case of new or worsening pulmonary symptoms, the administration of ADCETRIS should be discontinued during the evaluation until symptom relief.

Serious dermatological reactions: Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN), among them fatal, have been reported with ADCETRIS. If an SJS or TEN occurs, treatment with ADCETRIS should be discontinued and appropriate medical treatment initiated.

Gastrointestinal complications: ADCETRIS-treated patients have been reported to have acute pancreatitis, including fatalities. Other fatal and severe gastrointestinal complications including perforation, bleeding, erosions, ulcers, intestinal obstruction, enterocolitis, neutropenic colitis and ileus have been reported in ADCETRIS-treated patients. Lymphoma with pre-existing gastrointestinal involvement may increase the risk of perforation. If new or worsening gastrointestinal symptoms appear, a diagnostic evaluation should be made immediately and appropriate treatment should be initiated.

Embryo-fetal toxicity: Based on the mechanism of action and studies in animals, ADCETRIS may cause unborn life. Women of child-bearing potential are advised not to become pregnant during treatment with ADCETRIS and at least six months after the last dose of ADCETRIS. The most common (> 20%) adverse reactions: peripheral sensory neuropathy, fatigue, nausea, diarrhea, neutropenia, upper respiratory tract infection and fever.

Interaction with other medicinal products
Co-administration of potent CYP3A4 inhibitors or inducers and P-gp inhibitors may influence exposure to monomethylauristatin E (MMAE).

Use in special populations
Moderately or severely impaired hepatic function or severely impaired renal function: MMAE exposure and adverse reactions are increased. An application should be avoided.

Men with sex partners of child-bearing potential are advised to use effective contraceptive methods during treatment with ADCETRIS and at least six months after the last dose of ADCETRIS.

Patients should be advised to report pregnancy immediately and not to breastfeed while receiving ADCETRIS.

Other important safety information, including special warnings (BOXED WARNING), can be found in the full prescribing information for ADCETRIS at www.seattlegenetics.com or www.ADCETRIS.com .

About ICLUSIG (ponatinib) tablets

ICLUSIG is a kinase inhibitor targeting primarily BCR-ABL1, a pathologically altered tyrosine kinase expressed in chronic myeloid leukemia (CML) and Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph + ALL). ICLUSIG is a targeted cancer therapeutics engineered specifically to inhibit the activity of BCR-ABL1 and its mutations using a computer-aided and structure-based drug design platform. Not only does ICLUSIG target the native form of BCR-ABL1, but also therapy-resistant mutations, including the T315I mutation, which is the least responsive to any of the mutations. ICLUSIG is the only approved TKI,, This mutation has been linked to resistance to all other TKIs. ICLUSIG, which was fully approved by the FDA in November 2016, is also approved in the EU, Australia, Switzerland, Israel, Canada and Japan.

In the US, ICLUSIG is indexed to:

Treatment of adult patients with CML in the chronic, accelerated or blast phase (CP-CML, AP-CML or BP-CML) or Ph + ALL, in which no other tyrosine kinase inhibitor (TKI) is indicated.
Treatment of adult patients with T315I-positive CML (CP, AP or BP) or T315I-positive Ph + ALL.
Limitations of use: Treatment with ICLUSIG is not indicated and recommended for patients with newly diagnosed CP-CML.

IMPORTANT SAFETY INSTRUCTIONS FOR ICLUSIG (ponatinib) (USA)

WARNING: ARTERIAL CLOSURE, VENOUS THROMBOEMBOL, HEART FAILURE AND LIVER TOXICITY

The detailed Special Warnings (Boxed Warning) can be found in the full prescribing information.

At least 35 percent of ICLUSIG (ponatinib) -treated patients had arterial occlusions, including fatal myocardial infarction, stroke, large cerebral stenosis, severe peripheral vascular disease, and the need for emergency revascularization surgery. Patients with and without cardiovascular risk factors, including patients younger than 50, were affected by these events. If arterial occlusion occurs, treatment with ICLUSIG should be discontinued or discontinued immediately. The decision whether to resume treatment with ICLUSIG should be based on a risk-benefit assessment.
Venous thromboembolism occurred in 6 percent of ICLUSIG-treated patients. Patients should be monitored for signs of thromboembolism. In patients who develop severe venous thromboembolism, dose adjustment or discontinuation of ICLUSIG should be considered.
Heart failure, including fatal cases, was found in 9 percent of ICLUSIG-treated patients. Accordingly, the heart function is to be monitored. In the event of a new onset or worsening of heart failure, treatment with ICLUSIG should be discontinued or discontinued.
Hepatotoxicity, liver failure and death were observed in ICLUSIG-treated patients. Accordingly, liver function should be monitored. If liver toxicity is suspected, treatment with ICLUSIG should be discontinued.
WARNINGS AND PRECAUTIONS
Arterial occlusions:At least 35 percent of ICLUSIG-treated patients in Phase 1 and 2 trials had arterial occlusions, including fatal myocardial infarction, stroke, large cerebral stenosis, and severe peripheral artery disease. In the Phase 2 study, 33 percent (150/449) of ICLUSIG-treated patients had a cardiovascular (21%), peripheral (12%), or cerebrovascular (9%) arterial occlusive event; Some patients suffered more than 1 type of such event. Fatal and life-threatening events occurred within 2 weeks after initiation of therapy, even at doses as low as 15 mg per day. ICLUSIG may also cause recurrent or multi-site occlusion. Patients required revascularization procedures. The median time to onset of the first cardiovascular, cerebrovascular and peripheral vascular occlusive events was 193, 526 and 478 days, respectively. Patients with and without cardiovascular risk factors, including patients up to 50 years of age, were affected by these events. The most common risk factors associated with these events were hypertension, hyperlipidemia and history of heart disease. Arterial obstruction events increased with age and in patients with ischemia, hypertension, diabetes or hyperlipidemia.

Venous thromboembolism: Venous thromboembolic events occurred in 6 percent (25/449) of ICLUSIG-treated patients. Incidence rates were 5 percent (13/270 CP-CML), 4 percent (3/85 AP-CML), 10 percent (6/62 BP-CML), and 9 percent (3/32 Ph + ALL). Events included deep vein thrombosis, pulmonary embolism, superficial thrombophlebitis, and retinal vein thrombosis with vision loss. In patients who develop severe venous thromboembolism, dose adjustment or discontinuation of ICLUSIG should be considered.

Heart failure: 6% of ICLUSIG-treated patients (29/449) had fatal and severe cardiac insufficiency or left ventricular dysfunction. 9 percent of patients (39/449) had heart failure or left ventricular dysfunction of any grade. The most frequently reported cardiac insufficiency events were congestive heart failure and reduced ejection fraction (14 patients, 3% each). Patients should be monitored for signs and symptoms of heart failure and treated as clinically indicated, including discontinuation of ICLUSIG therapy. If severe heart failure develops, discontinuation of Iclusig should be considered

hepatotoxicity:ICLUSIG can cause liver toxicity, liver failure and death. Fulminant liver failure with fatal outcome occurred in one patient within one week of initiation of ICLUSIG treatment. Two more deaths from acute liver failure also occurred. The fatal cases occurred in patients with BP-CML or Ph + ALL. Heavy liver toxicity was observed in all disease cohorts, with 11 percent (50/449) showing grade 3 or 4 liver toxicity. The most common form of liver toxicity was elevations of AST or ALT (54% all grade, 8% grade 3 or 4, 5% not normalized at last follow-up), bilirubin and alkaline phosphatase. Hepatotoxic events were observed in 29 percent of patients. The median time to onset of the hepatic toxicity event was 3 months. Laboratory monitoring of liver function should be performed at baseline and then at least once a month or after clinical indication. Treatment with ICLUSIG should be discontinued, reduced or discontinued depending on the clinical judgment.

Arterial hypertension:Treatment-related increases in systolic or diastolic blood pressure (RR) occurred in 68 percent (306/449) of ICLUSIG-treated patients. 53 patients (12%) developed severe symptomatic blood pressure elevation, including hypertensive crisis, as serious adverse reactions. Patients whose blood pressure is associated with confusion, headache, chest pain or shortness of breath may require emergency clinical intervention. In patients with systolic RR <140 mm Hg and diastolic RR <90 mm Hg at baseline, 80% (229/285) had treatment-related hypertension, where 44 percent (124/285) developed stage 1 and 37 percent stage 2 hypertension. Of the 132 patients with stage 1 onset hypertension, 67 percent (88/132) developed stage 2 hypertension. During therapy with ICLUSIG, patients should be monitored for blood pressure elevations and, if necessary, treated until normalization of blood pressure is achieved. If hypertension can not be stopped by medication, treatment with ICLUSIG should be discontinued, reduced in dose or discontinued. In the event of marked deterioration and unstable or refractory hypertension, treatment should be discontinued and consideration given to the presence of renal artery stenosis should be considered.

pancreatitis:Pancreatitis occurred in 7% (31/449, 6% severe or grade 3/4) of ICLUSIG-treated patients. The incidence of treatment-related lipase elevations was 42 percent (16% at least grade 3). Because of pancreatitis, 6 percent of patients (26/449) discontinued or discontinued therapy. The median time to onset of pancreatitis was 14 days. In 23 of the 31 cases of pancreatitis, it receded within 2 weeks of discontinuation or dose reduction. Serum lipase should be monitored every two weeks for the first 2 months and then monthly or as clinically indicated. In patients with a history of pancreatitis or alcoholism, additional controls of serum lipase levels should be considered. Interruption of treatment or dose reduction may be required. In cases where lipase elevations are associated with abdominal symptoms, treatment with ICLUSIG should be discontinued and the patient should be screened for the presence of pancreatitis. Recovery of ICLUSIG treatment should not be considered until all symptoms have resolved and lipase levels have dropped to less than 1.5 times the upper limit of normal. If lipase elevations are associated with abdominal symptoms, discontinue treatment with ICLUSIG and examine the patient for the presence of pancreatitis. Recovery of ICLUSIG treatment should not be considered until all symptoms have resolved and lipase levels have dropped to less than 1.5 times the upper limit of normal. If lipase elevations are associated with abdominal symptoms, discontinue treatment with ICLUSIG and examine the patient for the presence of pancreatitis. Recovery of ICLUSIG treatment should not be considered until all symptoms have resolved and lipase levels have dropped to less than 1.5 times the upper limit of normal.

Increased toxicity of newly diagnosed CML in the chronic phase:In a prospective randomized clinical trial for first-line treatment of patients with newly diagnosed CML in the chronic phase (CP CML), monotherapy with ICLUSIG 45 mg once daily doubled the risk of serious adverse reactions compared to monotherapy with imatinib 400 mg once daily. Median exposure to treatment was less than 6 months. The study was stopped in October 2013 for safety reasons. Arterial and venous thrombosis and occlusion were at least twice as common in the ICLUSIG arm than in the imatinib arm. Compared with imatinib-treated patients, ICLUSIG-treated patients had a higher incidence of myelosuppression, pancreatitis, Liver toxicity, heart failure, hypertension and diseases of the skin and subcutaneous tissue. Treatment with ICLUSIG is not recommended and recommended for patients with newly diagnosed CP-CML.

neuropathy:Neuropathies of the peripheral nerves and cranial nerves were observed in patients treated with ICLUSIG. Overall, 20 percent (90/449) of ICLUSIG-treated patients had peripheral grade neuropathy (2%, grade 3/4). The most common reported peripheral neuropathies were paraesthesia (5%, 23/449), peripheral neuropathy (4%, 19/449), hypesthesia (3%, 15/449), dysgeusia (2%, 10/449), muscle weakness (2 %, 10/449) and hyperesthesia (1%, 5/449). Two percent (10/449) of ICLUSIG-treated patients (<1%, 3/449 – grade 3/4) developed cranial nerve neuropathy. Among the patients who developed neuropathy, this occurred in 26 percent (23/90) of cases in the first month of treatment. Patients should be monitored for the appearance of symptoms of neuropathy such as hypoaesthesia, hyperesthesia, paraesthesia, discomforting and burning sensations, neuropathic pain or weakness. If neuropathy is suspected, interruption of ICLUSIG treatment and clarification of symptoms should be considered.

Toxicity to the eye:Patients treated with ICLUSIG have been reported to have severe eye toxic effects leading to blindness or blurred vision. Retinal toxicity, including macular edema, retinal venous occlusion and retinal haemorrhage, was found in 2 percent of ICLUSIG-treated patients. Conjunctival irritation, corneal erosion, dry eye, conjunctivitis, conjunctival hemorrhage, hyperaemia and edema or eye pain occurred in 14 percent of patients. Blurred vision was reported by 6 percent of patients. Other toxic effects on the eye included cataract, periorbital edema, blepharitis, glaucoma, eyelid edema, iritis, iridocyclitis, and ulcerative keratitis.

Bleeding: 6 percent (28/449) of ICLUSIG-treated patients experienced major bleeding events, including fatal cases. Bleeding occurred in 28 percent (124/449) of the patients. Patients with AP-CML, BP-CML, and Ph + ALL had a higher incidence of major bleeding events. Gastrointestinal haemorrhage and subdural hematomas were the most frequently reported major bleeding events, occurring at 1 percent (4/449), respectively. Most, but not all, bleeding events occurred in patients with Grade 4 thrombocytopenia. Treatment with ICLUSIG should be discontinued in case of serious or severe bleeding and the cause of bleeding should be clarified.

Fluid retention: 4 percent (18/449) of the patients treated with ICLUSIG experienced severe fluid retention. A case of brain edema was deadly. Regarding fluid retention events, which occurred in> 2 percent of patients (treatment-related), including severe cases, pleural effusion (7/449, 2%), pericardial effusion (4/449, 1%), and peripheral edema (2/449, <1%).

Overall, fluid retention occurred in 31 percent of patients. The most common forms of fluid retention were peripheral edema (17%), pleural effusion (8%), pericardial effusion (4%), and peripheral swelling (3%).

Patients should be monitored for storage of fluid and treated as clinically indicated. Treatment with ICLUSIG should be discontinued, reduced or discontinued according to the clinical situation.

Cardiac arrhythmias: Arrhythmias occurred in 19 percent (86/449) of ICLUSIG-treated patients, with 7 percent (33/449) at least third-degree. Ventricular arrhythmias accounted for 3 percent (3/86) of all cardiac arrhythmias, with a grade of at least third grade. Symptomatic bradyarrhythmias leading to pacemaker implantation occurred in 1 percent (3/449) of ICLUSIG-treated patients.

Atrial fibrillation was the most common cardiac arrhythmia and occurred in 7 percent (31/449) of the patients, with about half of the third or. 4th degree were. Other third or fourth degree arrhythmia events included syncope (9 patients, 2.0%), tachycardia and bradycardia (2 patients, 0.4% each), and ECG QT prolongation, atrial flutter, supraventricular tachycardia, ventricular Tachycardia, atrial tachycardia, complete AV block, respiratory circulatory arrest, unconsciousness and sinus node dysfunction (1 patient each, 0.2%). 27 patients were hospitalized for the event.

In patients with symptoms of slow (fainting, dizziness) or rapid heart rate (chest pain, palpitations or dizziness), ICLUSIG treatment should be discontinued and symptoms clarified.

Myelosuppression: Myelosuppression has been reported as an adverse reaction in 59 percent (266/449) of ICLUSIG-treated patients, with grade 3 or 4 myelosuppression observed in 50 percent (226/449) of patients. The incidence of these events was higher in patients with AP-CML, BP-CML, and Ph + ALL than in patients with CP-CML. Severe myelosuppression (Grade 3 or 4) was observed at the early stages of treatment, with a median beginning of 1 month (range <1-40 months). In the first 3 months every 2 weeks, then monthly or as clinically indicated, create a large blood count and adjust the dose as recommended.

Tumor lysis syndrome: Two of the ICLUSIG-treated patients (<1%, one with AP-CML and one with BP-CML) developed a serious tumor lysis syndrome. Hyperuricemia was found in 7 percent (31/449) of the patients. Since tumor lysis syndrome may be present in patients with advanced disease, adequate hydration and treatment with elevated uric acid levels should be considered prior to initiation of ICLUSIG therapy.

Reversible Posterior Leukoencephalopathy Syndrome (RPLS): Cases of reversible posterior leukoencephalopathy syndrome (RPLS – also known as posterior reversible encephalopathy syndrome (PRES)) have been reported in the post-marketing surveillance study in ICLUSIG-treated patients. RPLS is a neurological disorder characterized by symptoms such as seizure, headache, decreased attention, altered mental functions, vision loss, and other neurological and visual disturbances. Often there is hypertension and the diagnosis is made with supportive brain magnetic resonance imaging (MRI) findings. If the RPLS diagnosis is made, treatment with ICLUSIG should be discontinued. Therapy should only be restarted

Impairment of wound healing and gastrointestinal perforation: Since ICLUSIG may interfere with wound healing, treatment with ICLUSIG should be discontinued at least one week prior to major surgery. In one patient, 38 days after cholecystectomy, a severe gastrointestinal perforation (fistula) occurred.

Embryo-fetal toxicity: The mechanism of action and observations in animals suggest that ICLUSIG may induce fetal harm when used in pregnancy. In animal reproduction studies, oral administration of ponatinib to pregnant rats during the organogenesis phase caused adverse developmental effects at exposure lower than human exposure at the human recommended dose. Pregnant women should be made aware of the potential risk to the unborn child. Women of childbearing potential should use effective contraception during treatment with ICLUSIG and during the 3 weeks following the last dose.

INACCURATE RESPONSES
Most common adverse reactions: Overall, the most common non-hematological adverse reactions (≥20%) were abdominal pain, rash, constipation, headache, dry skin, arterial occlusion, fatigue, hypertension, fever, arthralgia, nausea, diarrhea, lipase elevation, vomiting, muscle pain and pain in one limb. Hematologic adverse reactions included thrombocytopenia, anemia, neutropenia, lymphopenia and leukopenia.

To report POSSIBLE REACTIONS, contact Takeda at 1-844-T-1POINT (1-844-817-6468) or the FDA at 1-800-FDA-1088 or www.fda.gov/medwatch .

INTERACTIONS WITH OTHER MEDICINES
Strong CYP3A inhibitors: Avoid concomitant use or reduce the dose of ICLUSIG if co-administration can not be avoided.
Strong CYP3A inducers: avoid concomitant use.

Use in special groups of patients
Women of childbearing potential and fertile men: Use of ICLUSIG during pregnancy may cause damage to the unborn child. Women are advised to use effective contraception during treatment with ICLUSIG and in the 3 weeks following the last dose. Ponatinib may affect fertility in women, and it is not known if these effects are reversible. In women of childbearing potential, pregnancy status should be checked before initiating therapy with ICLUSIG.

Breast-feeding: Women are advised not to breastfeed during ICLUSIG therapy and six days after the last dose.

The US prescribing information can be found at: View Source

About NINLARO (ixazomib) capsules

NINLARO (ixazomib) is an oral proteasome inhibitor that is also being studied in the multiple myeloma therapy continuum and in systemic light chain amyloidosis (AL). It was the first oral proteasome inhibitor studied in Phase 3 clinical trials and received approval. NINLARO was approved by the US Food and Drug Administration in November 2015 following a Priority Review and by the European Commission in November 2016. In the US and Europe, NINLARO in combination with lenalidomide and dexamethasone is indicated for the treatment of patients with multiple myeloma who have received at least one prior therapy. NINLARO has received regulatory approval from regulators in more than 55 countries.

Ixazomib has been granted orphan drug status in multiple myeloma in the United States and Europe in 2011 and in AL and amyloidosis in 2012 in the United States and Europe. Ixazomib was awarded FDA Breakthrough Therapy status in 2014 for relapsed or refractory systemic light chain amyloidosis (AL), a related extremely rare disease. The Japanese Ministry of Health, Labor and Social Affairs granted Ixazomib orphan drug status in 2016.

TOURMALINE, the comprehensive Ixazomib clinical development program, includes a total of six ongoing regulatory trials – five investigating all major groups of multiple myeloma patients and one dealing with light chain amyloidosis:

TOURMALINE-MM1 for testing ixazomib over placebo in combination with lenalidomide and dexamethasone in relapsed and / or refractory multiple myeloma.
TOURMALINE-MM2 for the testing of ixazomib versus placebo in combination with lenalidomide and dexamethasone in patients with newly diagnosed multiple myeloma.
TOURMALINE-MM3 for testing ixazomib versus placebo as a maintenance treatment in patients with newly diagnosed multiple myeloma after induction therapy and autologous stem cell transplantation (ASCT)
TOURMALINE-MM4 for testing ixazomib over placebo as maintenance treatment in patients with newly diagnosed multiple myeloma who have not undergone ASCT. Patients are currently enrolled in this study.
TOURMALINE-MM5 for the testing of ixazomib plus dexamethasone versus pomalidomide plus dexamethasone in patients with relapsed and / or refractory multiple myeloma who have become resistant to lenalidomide.
TOURMALINE-AL1 for the testing of ixazomib plus dexamethasone for a physician on the basis of a selection of therapies in patients with relapsed or refractory AL amyloidosis. Patients are currently enrolled in this study.
For more information on actively recruiting Phase 3 studies, visit View Source

In addition to the TOURMALINE study program, Ixazomib is currently being evaluated in several therapeutic combinations in various patient populations worldwide through investigator-initiated studies (IIT).

NINLARO (Ixazomib) Capsules: Important Safety Information Worldwide

SPECIAL WARNING (S) AND PRECAUTIONS
Thrombocytopenia been reported with NINLARO (28% vs 14% with the NINLARO or placebo regimen). Platelets reached their lowest point between the 14th and 21st day of the 28-day treatment cycle and recovered to baseline by the start of the next cycle. This did not lead to increased bleeding events or platelet transfusions. During treatment with NINLARO, the platelet count should be monitored at least monthly and more frequent monitoring should be considered during the first three cycles. Thrombocytopenia should be treated with dose adjustment and platelet transfusions in accordance with the recommendations of the standard guidelines.

Gastrointestinal toxicities were reported in the NINLARO and placebo regimens, for example, diarrhea (42% vs. 36%), constipation (34% vs. 25%), nausea (26% vs. 21%), and vomiting (22% vs 11%). These occasionally required the use of medication for vomiting and diarrhea, as well as supportive therapy.

Peripheral neuropathy has been reported in NINLARO (28% vs. 21% with the NINLARO or placebo regimen). The most commonly reported adverse reaction was peripheral sensory neuropathy (19% and 14% in the NINLARO or placebo regimen). Peripheral motor neuropathy has not been reported in any of the two regimens (<1%). Patients should be monitored for signs of peripheral neuropathy and dosing adjusted if necessary.

Peripheral edema has been reported in NINLARO (25% vs. 18% with the NINLARO or placebo regimen). The underlying causes should be clarified. If necessary, patients should receive supportive care. Dose adjustment should be made with dexamethasone according to the SPC or with NINLARO if severe symptoms occur.

Skin reactions occurred in 19 percent of patients with the NINLARO regimen compared to 11 percent of patients on the placebo regimen. The most common form of rash on both schemes was a maculopapular and macular rash. Rashes should be treated with supportive therapy, dose adjustment or discontinuation of the drug

Hepatotoxicity, drug-induced liver damage, hepatocellular damage, fatty liver, and cholestatic hepatitis have not been reported frequently in NINLARO-treated patients. Liver values ​​should be monitored regularly and dose adjustments should be made for symptoms of Grade 3 and 4.

Pregnancy – NINLARO can lead to harm to unborn life. For fertile men and women of childbearing potential, contraceptive methods should be used during treatment and for a further 90 days after the last dose of NINLARO. Due to the possible risk to the unborn child, women of childbearing age should be prevented from taking pregnancy while being treated with NINLARO. Women using hormonal contraceptives should also use a barrier prevention method.

Breastfeeding – It is not known whether NINLARO or its breakdown products are excreted in breast milk. Due to possible adverse events in breast-fed infants, NINLARO-treated patients should abstain.

SPECIAL PATIENT POPULATIONS
Hepatic impairment: The starting dose of NINLARO should be reduced to 3 mg in patients with moderate or severe hepatic impairment.

Renal impairment: In patients with severe renal insufficiency or patients with end-stage renal disease (ESRD) requiring dialysis, the starting dose of NINLARO should be reduced to 3 mg. NINLARO is not dialyzable and can therefore be administered independently of the dialysis schedule.

INTERACTIONS WITH OTHER MEDICINES
The simultaneous use of NINLARO and potent CYP3A inducers is foreseeable.

UNWANTED REACTIONS
The most common adverse reactions seen in at least 20% of patients treated with the NINLARO regimen or more frequently than placebo regimens were diarrhea (42% vs. 36%), constipation (34% vs. 25%). ), Thrombocytopenia (28% vs. 14%), peripheral neuropathy (28% vs. 21%), nausea (26% vs. 21%), peripheral edema (25% vs. 18%), vomiting (22% vs. 11%) and back pain (21% vs. 16%). The severe adverse reactions that occurred in at least 2 percent of patients included thrombocytopenia (2%) and diarrhea (2%). For each adverse reaction, one or more of the three drugs was discontinued in ≤ 1 percent of patients in the NINLARO regimen.

The EU Prescribing Information can be found at View Source
The US prescribing information can be found at https: //www.ninlarohcp .com / pdf / prescribing-information.pdf
You can find the Canadian product monograph at View Source

Xencor Reports Third Quarter 2018 Financial Results

On November 5, 2018 Xencor, Inc. (NASDAQ: XNCR), a clinical-stage biopharmaceutical company developing engineered monoclonal antibodies for the treatment of autoimmune disease, asthma and allergic diseases, and cancer, reported financial results for the third quarter ended September 30, 2018 and provided a review of recent business and clinical highlights (Press release, Xencor, NOV 5, 2018, View Source [SID1234530746]).

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!

"Our recent data readouts represent advancement across our pipeline of wholly owned and partnered XmAb-based therapeutics for autoimmune disorders and cancer," said Bassil Dahiyat, Ph.D., president and chief executive officer at Xencor. "In particular, we are encouraged by initial data from the ongoing Phase 1 study of our lead bispecific oncology candidate, XmAb14045, in AML, which will be presented in an oral session at ASH (Free ASH Whitepaper) next month. These data show complete remissions on a weekly dosing schedule in heavily pretreated patients as we continue to optimize dosing regimen."

Dr. Dahiyat added, "This is the first clinical data to emerge from our bispecific oncology programs and reflects the potential of our novel bispecific Fc domains to enable stable, long-lived bispecific antibodies in which their potencies are tuned to potentially improve tolerability and effectiveness. Our broad pipeline now includes seven bispecific candidates in addition to our lead autoimmune disease candidate, XmAb5871, which is expected to enter into a Phase 3 study in IgG4-RD by early 2019."

Recent Business Highlights and Upcoming Clinical Plans

XmAb5871: XmAb5871 is a first-in-class monoclonal antibody that targets CD19 with its variable domain and uses Xencor’s XmAb immune inhibitor Fc domain to target FcγRIIb, a receptor that inhibits B-cell function. XmAb5871 is currently in clinical development for IgG4-Related Disease (IgG4-RD) and Systemic Lupus Erythematosus (SLE), and it has received Orphan Drug designation from the FDA and Orphan Medicinal Product designation from the European Commission for the treatment of IgG4-RD.

Based on promising Phase 2 results and ongoing discussions with the regulatory authorities, Xencor is designing a randomized, placebo-controlled, double-blind Phase 3 trial of XmAb5871 in approximately 200 to 250 patients and is defining the novel endpoint in order to evaluate the addition of XmAb5871 to standard of care. Initiation of the study is expected by early 2019.
In October 2018, Xencor presented topline results from its randomized, double-blind, placebo-controlled Phase 2 study in patients with SLE at the American College of Rheumatology (ACR) Annual Meeting. A positive trend was observed in the primary endpoint of the study, proportion of efficacy-evaluable patients who did not experience loss of improvement (LOI) by Day 225, though it did not achieve statistical significance. The study achieved the prespecified secondary endpoint, time to LOI, and patients treated with XmAb5871 experienced a 76% improvement in median time to LOI compared to patients treated with placebo. Given these encouraging results, Xencor believes that XmAb5871 warrants further development in SLE and is seeking a partner to continue such development.

Bispecific Oncology Pipeline: Xencor’s bispecific Fc domains are being used to develop several classes of novel drug candidates, including: CD3 bispecific antibodies, tumor microenvironment (TME) activator bispecific antibodies and bispecific cytokines. Xencor’s XmAb Fc domains confer long circulating half-lives, stability and ease of manufacture.

CD3 Bispecific Antibodies: Xencor’s initial bispecific antibody programs are tumor-targeted antibodies that contain both a tumor antigen binding domain and a cytotoxic T-cell binding domain (CD3). These bispecific antibodies activate T cells for highly potent and targeted killing of malignant cells.

Presentation of initial data from Phase 1 study of XmAb14045 (CD123 x CD3) in patients with acute myeloid leukemia (AML) on December 3, 2018 at the American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting.
Initial data from Phase 1 study of XmAb13676 (CD20 x CD3) in B-cell malignancies, expected in 2019, pending alignment on timing with Novartis.
Initial data from Phase 1 study of XmAb18087 (SSTR2 x CD3) in neuroendocrine tumors and gastrointestinal stromal tumors, expected in 2019.
TME Activator Bispecific Antibodies: Xencor’s bispecific pipeline includes a suite of TME activators that engage multiple targets, such as T-cell checkpoints or agonists.

Initial data from DUET-2, a Phase 1 study of XmAb20717 (PD-1 x CTLA-4) in advanced solid tumors, expected in 2019.
IND application for XmAb23104 (PD-1 x ICOS) allowed by the FDA in November 2018; initiation of Phase 1 study in select solid tumors expected in 2019.
IND submission for XmAb22841 (CTLA-4 x LAG-3) in multiple oncology indications, expected by year-end 2018; initiation of Phase 1 trial expected in 2019.
Bispecific Cytokines: Xencor is developing a candidate that contains cytokine and cytokine receptor domains to selectively expand and activate immune cells that can be recruited against tumors.

IND submission for XmAb24306 (IL15/IL15Rα-Fc) in multiple oncology indications expected in 2019.
XmAb7195: XmAb7195 is a first-in-class monoclonal antibody that targets IgE with its variable domain and uses Xencor’s XmAb immune inhibitor Fc domain to target FcyRIIb, resulting in three distinct mechanisms of action for reducing IgE. In a Phase 1b study, subcutaneously-administered XmAb7195 induced potent IgE reduction with improved tolerability. Xencor is currently seeking a development partner for XmAb7195.

Partnered XmAb Programs: Eight pharmaceutical companies and the National Institutes of Health are advancing novel drug candidates either discovered at Xencor or that rely on Xencor’s proprietary XmAb technology. Four such programs are currently undergoing clinical testing, including MOR208, which is in Phase 3 development as a combination agent for the treatment of relapsed or refractory diffuse large B-cell lymphoma, and AMG 424, a CD38 x CD3 bispecific antibody, which Amgen announced had entered into a Phase 1 study for the treatment of patients with multiple myeloma in the third quarter of 2018.

In the third quarter of 2018, Xencor received $9 million in milestone payments from Alexion in connection with their submission of marketing authorizations for ALXN1210 to the FDA and EMA for the treatment of patients with paroxysmal nocturnal hemoglobinuria. In October 2018, Alexion announced that they had submitted a marketing authorization to regulatory authorities in Japan and that the FDA had set the review date for its application for February 2019.

Third Quarter Ended September 30, 2018 Financial Results

Effective January 1, 2018, Xencor adopted the new revenue recognition standard, Accounting Standard Codification 606 (ASC 606). In addition to adopting the standard for 2018, revenue reported for the prior period ending September 30, 2017 has been revised to reflect the new standard.

Cash, cash equivalents and marketable securities totaled $547.8 million as of September 30, 2018, compared to $363.3 million at December 31, 2017. The increase reflects net proceeds of $245.5 million from Xencor’s sale of additional stock in March 2018, partially offset by cash used to fund operating activities in the nine months ended September 30, 2018.

Total revenue for the three- and nine-month periods ended September 30, 2018 was $29 million, compared to zero and $16 million of revenue reported for the same periods in 2017. Revenues in the three and nine-month periods ended September 30, 2018 included revenue recognized under the Company’s Novartis collaboration and milestone payments received from the Company’s Alexion collaboration.

Research and development expenditures for the third quarter ended September 30, 2018 were $21.0 million, compared to $19.4 million for the same period in 2017. Total research and development expenditures for the nine-month period ended September 30, 2018 were $70.4 million, compared to $51.4 million for the same period in 2017. The increased research and development spending for the three and nine months ended September 30, 2018 reflects additional spending on Xencor’s expanding pipeline of bispecific oncology candidates.

General and administrative expenses for the third quarter ended September 30, 2018 were $7.4 million, compared to $4.2 million in the same period in 2017. Total general and administrative expenditures for the nine-month period ended September 30, 2018 were $17.0 million, compared to $13.1 million for the same period in 2017. The increased spending on general and administrative expenses for the three and nine months ended September 30, 2018 reflects increased compensation costs including increased stock-based compensation charges.

Non-cash, stock-based compensation expense for the nine months ended September 30, 2018 was $15.5 million, compared to $10.2 million for same period in 2017.

Net income for the third quarter ended September 30, 2018 was $3.2 million, or $0.05 on a fully diluted per share basis, compared to a net loss of $22.7 million, or $(0.48) on a fully diluted per share basis, for the same period in 2017. The net income reported for three months ended September 30, 2018 over the loss for the same period in 2017 is primarily due to revenue recognized from Xencor’s Novartis and Alexion collaborations in 2018. For the nine months ended September 30, 2018, net loss was $52.2 million, or $(0.98) on a fully diluted per share basis, compared to a net loss of $45.9 million, which was also $(0.98) on a fully diluted per share basis, for the same period in 2017. The increased revenue for the nine months ended September 30, 2018 over amounts for the same period in 2017 was offset by increased spending in research and development in 2018. The earnings per share loss for the nine months ended September 2018 was equal to the earnings per share loss in 2017 due to the increase in shares outstanding in 2018.

The total shares outstanding were 56,212,449 as of September 30, 2018, compared to 46,955,365 as of September 30, 2017. The additional shares outstanding at September 30, 2018 reflect the 8,395,000 shares sold in Xencor’s March financing.

Financial Guidance

Based on current operating plans, Xencor expects to have cash to fund research and development programs and operations into 2023. Xencor expects to end 2018 with approximately $525 million in cash, cash equivalents and marketable securities.

Conference Call and Webcast

Xencor will host a conference call today at 4:30 p.m. ET (1:30 p.m. PT) to discuss these third quarter 2018 financial results and provide a corporate update.

The live call may be accessed by dialing (877) 359-9508 for domestic callers or (224) 357-2393 for international callers and referencing conference ID number 5577136. A live webcast of the conference call will be available online from the Investors section of the Company’s website at www.xencor.com. The webcast will be archived on the company’s website for 90 days.