Friday, May 22, 2015

blue 赛百诺内斗 创新成功却市场失败是中国生物制药领域的通病


Biotech, healthcare, value, research analyst
Profile| Send Message|
( followers)
| Performance

                    

科学家大战资本家:基因治疗药物“今又生”之殇

创新成功为何败走市场?

单纯抛开赛百诺创始人和控股方老板的个性问题,赛百诺内斗依然值得反思,为何创新成功却在市场失败?
中国社科院《科学新闻》原总编辑贾鹤鹏一直关注生物制药科技创新,他认为,“今又生”以及其他中国创新药(安柯瑞、恩度)的经历证明,这种创新成功却市场失败是中国生物制药领域的通病。“做出了在科学上领先世界的新药研发的科学家大部分都离职了。离职的原因各异,但都是与资方(包括国企)合不来。”
为何会这样?贾鹤鹏认为,客观环境而言,整个体制没有做好承接真正创新药的准备。
“这个体制可以通过绿色通道和科研经费支持让真正有能力的科学家走出科学上的成功,但是,首先不能以医保覆盖来支持新药的推广,其次不能提供一个环境让真正的创新药战胜包括医疗腐败和医患关系(导致医生不敢用新药,不敢尝试新疗法)这些体制性因素,第三不能形成行业的良性互动(比如由真正有实力又有能力的大型药企兼并赛百诺)。”
“而在资金方面,研发时没有风险投资,直到现在仍然主要靠国家科研经费来缓解,所谓风险投资都是等拿到了新药证书,而到了这个阶段,创新企业其实需要的不是业外的财务投资,而是业内的投资。”
一名熟悉赛百诺的香港投资咨询公司负责人分析,对投资人大股东奔达的运作而言,有许多方面值得反思和商榷。
首先,它应该在受让股权进入赛百诺前弄清楚赛百诺的资产状况,包括无形资产的法律状况,以及原股东及管理层与公司的各种关系,这样就完全可以避免上述各种诉讼的发生,股东及管理层就可以齐心协力发展公司业务,使股东权益最大化。其次,它应该遵循高科技企业的发展规律,充分尊重科技人员的贡献和努力,而不是将其按一般企业管理和运营,使赛百诺失去研发后劲及技术含量而沦为普通企业。再次,投资人应该摒弃民营企业家宁为鸡头不为凤尾的落后观念,吸收对公司发展有决定意义的资金和专业人员,共同发展
                                                                   

生物圈内所有内容对注册用户免费开放!请加入我们,同筑纯净的生物科研学术交流平台。

您需要 登录 才可以下载或查看,没有帐号?加入乐研 qq_login
x
科学家大战资本家:基因治疗药物“今又生”之殇
彭朝晖与徐卫是深圳市赛百诺基因技术有限公司(简称赛百诺)的创始人和现任总裁。过去8年,双方一直在中国法庭上争斗。
命运把双方纠缠在一起。1998年,“海归”彭朝晖在深圳成立赛百诺,研发试用的世界首个基因治疗药物“今又生”于2003年获批上市。2006年,徐卫的公司通过股权转让方式控股了赛百诺,并于2008年通过赛百诺与彭朝晖就5项中国发明专利和3项国际发明专利(已在36个国家授权)的专利权属打起了官司。
官司打了8年,从深圳市中院的一审到广东省高院的二审,两次判决都认定赛百诺胜诉。彭朝晖不服提出抗诉申请,经省高检、最高检各级程序抗诉,最高法立案裁定指令广东省高院再审,中止二审原判决的执行。3月24日,在广东省高院的法庭上,双方内斗迎来最后一搏。
经8年纠纷,双方皆双鬓染霜。此案虽然尚未宣判,但赛百诺董事万宜青承认,此事没有赢家,8年诉讼,企业也受到重创,“今又生”更难完善,甚至变得默默无闻。
为何会这样?广东推出创新驱动发展决策,赛百诺的发展经历为科技企业创新发展带来深重教训。

融资不成引来日后对手

1998年初,“海归博士”彭朝晖在深圳成立赛百诺公司,主导了“重组腺病毒-p53抗癌注射液”(注册商标名:“今又生”/Gendicine)的工艺优化、临床试验。2003年,“今又生”获得国家批准,成为世界上第一个获准上市的基因治疗药,引起国内外主流媒体和权威医学机构聚焦,彭朝晖也一时成为耀眼的科技明星。
声名鹊起之时,彭朝晖从实验室走向市场,扩建厂房、引进人才、扩大临床试验等各项投入骤增,却没有达到预期销售额,主要资金支持方政府及国企又相继停止投入,一时令公司陷入了财务困境,只懂科研的彭朝晖开始四处融资,但频遭不顺。
与此同时,彭朝晖未来的老对手徐卫也在发愁。
据徐卫介绍,其由一名医生下海,白手起家。由于没有核心知识产权,产品主要以原料药和维生素为主,湖北同济奔达鄂北制药公司(简称奔达)始终摆脱不了数量大、利润低的阴影。困难的市场环境下,万宜青、徐卫夫妇练就其特别重的“狼性”。为了转型升级,徐卫将奔达送到美国去借壳上市,还走过弯路,“非常痛苦”。上市后,徐卫计划募集资金去购买具备创新力的雏形企业。
这时,经营困难的赛百诺进入徐卫视线。
由于两个股东急于脱手,奔达由本欲增资获得代理权,迅速转变为向赛百诺原法人股东购买老股,2006年10月,一跃而成该公司大股东。注入高科技概念后,奔达在美国场外柜台交易系统(OTCBB)上市的股价因超额认购,赚得盆满钵满。彭朝晖似乎也看到了赛百诺国际化的路径。
一名要求匿名的投资界知情人士分析,因为过于草率,急于求成,赛百诺此阶段错误地引进了不能给公司带来用于发展的增量资金的股东,同时还给公司带来了极其不同的发展理念,为日后股东争执埋下隐患。
好景不长,奔达与彭朝晖在融资、股权和管理等问题上的矛盾升级。

“科学家”大战“资本家”

根据双方此前签名达成的合作共识,赛百诺拟成立医药、基因技术和研究所三马齐驱的公司治理组织结构框架,但奔达控股后并没有履行这一共识。
股权变更后,赛百诺董事会中只有万宜青、徐卫和彭朝晖三人,且万、徐为夫妇,掌握绝对控制权。同时,彭是法人代表和董事长,而徐是总裁,董事会成员和经营层成员重叠较多,治理结构存在明显问题,双方职责很容易模糊。
让彭朝晖无法接受的是,徐卫与万宜青多次在彭朝晖缺席的情况下做出公司重要决议。万宜青对此表示:“董事会就三人,你彭朝晖不来,那我们俩就通过了。”
内斗焦点为公司基因治疗相关专利权属。
2007年底,徐卫等人要求彭朝晖将专利权属无条件变更给已被其控制的赛百诺。彭朝晖提出,为了公司发展可协商,但要按此前对一份案外专利的做法,先通过对专利评估作价。
2008年8月,赛百诺向深圳市中院正式起诉彭朝晖等人的上述专利为职务发明,要求变更权属人为赛百诺。如果参考案外一个专利评估作价4000多万算,当时业内估算此案涉案专利标的额约达2亿多,这在国内极少见。
对于创新公司而言,有足够知本的研发团队,要在市场生存发展,如鱼一般,需资本的水,知本、资本相融方可共赢。从一审、二审到现在再审,双方缠斗了8年。媒体形容为“科学家VS资本家”。双方焦点集中在彭朝晖是否早期已经以涉案专利入股,涉案专利是否属于赛百诺的职务发明。
8年后的现在,彭朝晖回忆,奔达表示将会成为赛百诺资金后盾,而其在美国虽然不是正式上市,但也是柜台交易公司,而且其在赛百诺公司内部的公开场所摆放的专利材料已经明确显示专利权属彭朝晖等人。在一系列乐观心态驱使下,彭朝晖称犯了一个让他悔恨终身的错误——融资时没有对奔达做好尽职调查。而奔达方也“委屈”表示没有对对方进行尽职调查。
正因为此,甚至第二股东彭朝晖也被赛百诺拒之门外,其科研团队也被迫黯然离开,赛百诺此前引发世界关注的科研遂蒙尘多年。
动荡之下,“今又生”一度被国家食品药品监督管理局吊销了GMP证书,一年多以后才恢复。而此前政府投入1450万的“今又生国家高新技术产业示范工程”中断。

“早产儿”的遗憾

一出生便不同凡响,但只是“早产儿”,这是内斗给“今又生”带来的最大影响。
作为世界上首个被批准上市的基因治疗药物,“今又生”上市后曾被质疑缺乏有足够的临床试验数据来确证其抗癌效果,但也有科学证明其有效性。
2009年,当时北京肿瘤医院张珊文教授和福建省肿瘤医院潘建基教授及其同事,在各自医院共招募了82名鼻咽癌患者,随机分为两组,其中42名患者联合使用“今又生”和放疗;另外40名则作为对照,只进行放疗。
上述研究结果出现在《临床肿瘤学杂志》报告中,“今又生”组的五年生存率比对照组高出7.5%。“今又生”组的五年无病生存率比对照组也要高出约11.7%。
生物医药产业是个高投入、高风险的领域,新药的开发成功率约为万分之一,通常一个药从开发到顺利产业化至少要花12年,很少有风险投资能支持这样长期、巨额的投入。
2008年中国生物产业大会网站的资料显示,“今又生”在临床试验和应用中已经治疗约1万例癌症患者。这当中,还有约500名外国患者专程到中国接受治疗。
然而,因为核心科研人才流失,“今又生”药效难以改善,彭朝晖不得不承认是“早产儿”,但也不服气。
“早产儿,毕竟是儿,不是被堕胎或引产的胚胎。”彭朝晖日前就上述疑问回函称,对于三期临床试验数据少问题,“今又生”是在1998年12月被SDA(当时称中国药品监督管理局)批准进行临床试验,随后的临床试验沿袭当时的临床试验规范(GCP),即I期试验安全性,II期试验有效性,I、II期试验通过后即可批准生产,III期临床试验是药品上市以后的过程。国家批准的临床试验病种是“头颈部鳞癌”,包括20余种恶性肿瘤。上述论文发表的文章仅总结和5年随访了“头颈部鳞癌”之一鼻咽癌,实际做的病人数要大大多于文章发表的数量。

创新成功为何败走市场?

单纯抛开赛百诺创始人和控股方老板的个性问题,赛百诺内斗依然值得反思,为何创新成功却在市场失败?
中国社科院《科学新闻》原总编辑贾鹤鹏一直关注生物制药科技创新,他认为,“今又生”以及其他中国创新药(安柯瑞、恩度)的经历证明,这种创新成功却市场失败是中国生物制药领域的通病。“做出了在科学上领先世界的新药研发的科学家大部分都离职了。离职的原因各异,但都是与资方(包括国企)合不来。”
为何会这样?贾鹤鹏认为,客观环境而言,整个体制没有做好承接真正创新药的准备。
“这个体制可以通过绿色通道和科研经费支持让真正有能力的科学家走出科学上的成功,但是,首先不能以医保覆盖来支持新药的推广,其次不能提供一个环境让真正的创新药战胜包括医疗腐败和医患关系(导致医生不敢用新药,不敢尝试新疗法)这些体制性因素,第三不能形成行业的良性互动(比如由真正有实力又有能力的大型药企兼并赛百诺)。”
“而在资金方面,研发时没有风险投资,直到现在仍然主要靠国家科研经费来缓解,所谓风险投资都是等拿到了新药证书,而到了这个阶段,创新企业其实需要的不是业外的财务投资,而是业内的投资。”
一名熟悉赛百诺的香港投资咨询公司负责人分析,对投资人大股东奔达的运作而言,有许多方面值得反思和商榷。
首先,它应该在受让股权进入赛百诺前弄清楚赛百诺的资产状况,包括无形资产的法律状况,以及原股东及管理层与公司的各种关系,这样就完全可以避免上述各种诉讼的发生,股东及管理层就可以齐心协力发展公司业务,使股东权益最大化。其次,它应该遵循高科技企业的发展规律,充分尊重科技人员的贡献和努力,而不是将其按一般企业管理和运营,使赛百诺失去研发后劲及技术含量而沦为普通企业。再次,投资人应该摒弃民营企业家宁为鸡头不为凤尾的落后观念,吸收对公司发展有决定意义的资金和专业人员,共同发展
Cambridge, Massachusetts-based bluebird bio (NASDAQ:BLUE) is a $710M market cap clinical-stage biotechnology company that is developing gene therapies for severe genetic and orphan diseases.
Founded in 1992, the company was formerly known as Genetix Pharmaceuticals, Inc. The company's name was changed to bluebird bio, Inc. in September 2010. In addition to its headquarters in Cambridge, bluebird bio has operations in San Francisco, California, and Paris, France.

Pipeline

bluebird bio's pipeline includes:
  • Lenti-D to treat patients with childhood cerebral adrenoleukodystrophy (CCALD), a hereditary neurological disorder, as well as for the treatment of adult cerebral adrenoleukodystrophy (ALD);
  • LentiGlobin vector for the treatment of ß-thalassemia, a hereditary blood disorder; and sickle cell diseases (SCD); and
  • A preclinical oncology program in the chimeric antigen receptor T cells (CAR T) field under a collaboration with Celgene Corporation (NASDAQ:CELG).

Gene Therapy Platform and Process

Gene therapy is cutting edge science that strives to introduce a functional copy of a defective gene into a patient's cells through the gene transfer process. In this process, a functional gene is delivered and incorporated into a patient's cells through a delivery system called a vector. These vectors are usually based on viruses that have been altered to utilize the virus's natural ability to introduce genes into cells, but are modified to be non-replicating by deleting the part of the viral genome responsible for replication and disease.
bluebird bio's gene therapy platform is based on viral vectors that utilize a modified, non-replicating version of the human immunodeficiency virus type 1 (HIV-1), that has been stripped of all of the components necessary for it to mutate and infect additional cells. Since HIV-1 is part of the lentivirus family of viruses, the company refers to its vectors as lentiviral vectors. bluebird bio's lentiviral vectors are used to introduce a functional copy of a gene to a patient's hematopoietic stem cells (HSCs). HSCs are capable of differentiating into many cell types. Since HSCs are dividing cells, bluebird bio scientists have found that this approach allows for sustained expression of the modified gene because it takes advantage of the lifetime replication of gene-modified HSCs.
bluebird bio has also developed a proprietary cell-based vector manufacturing process. The company believes its innovations in viral vector design and manufacturing processes are important steps towards advancing the field of gene therapy to a commercial scale.
Although its initial focus is in childhood cerebral adrenoleukodystrophy (CCALD), ß-thalassemia and sickle cell disease (SCD), bluebird bio believes its gene therapy platform has broad therapeutic potential for many indications.
For beta-thalassemia, SCD, and adrenoleukodystrophy, a functional copy of the malfunctioning gene is inserted into a patient's HSCs, with the goal of genetically modifying a patient's cells to correct or address the genetic basis underlying the disease.
In oncology, the gene therapy process targets T cells. In these cases, genetic sequences are inserted into a patient's own T cells, which are intended to program the T cells to recognize and attack cancer cells. T cells, also known as lymphocytes, are a type of white blood cell that help the body fight diseases.
In October 2003, Shenzhen SiBiono GeneTech made history by becoming the first company approved to market a gene therapy. China's State Food and Drug Administration approved Genedicine for treatment of head and neck squamous cell carcinoma. but the company believes that continued clinical trials will prove Genedicine to be a safe and effective treatment for other types of cancer.
On November 2, 2012, the European Commission approved Uniqure's Glybera (alipogene tiparvovec), for the treatment of lipoprotein lipase deficiency. Glybera is the first gene therapy approved in the Western world. The treatment costs approximately $1.6 million per patient. Uniqure, a small Dutch company, has a product pipeline with several gene therapies for hemophilia B, acute intermittent porphyria, Parkinson's disease and Sanfilippo B. The Amsterdam, Netherlands-based company plans to meet with the FDA with the aim of a U.S. launch in 2014.
According to the industry intelligence advisor, RNCOS, the global gene therapy market is estimated to have reached approximately $485 million in 2011, and is forecast to grow significantly with the introduction of new products in developed countries. The research firm predicts the gene therapy market reach the mark of $650 million by the end of 2015.
RNCOS noted that adenovirus vectors hold a leading position in the ongoing gene therapy clinical studies. Its usage rate is around 24% in the gene therapy treatment trials being pursued by industry participants.
Companies, such as Advantagene, Applied Genetic Technologies, Ceregene, Sanofi/Genzyme (NYSE:SNY), GenVec (NASDAQ:GNVC), are conducting trials for gene therapy-based products using adenovirus vectors.
Other companies developing gene therapies include AnGes MG Inc., ANI Pharmaceuticals, Inc./BioSante Pharmaceuticals (NASDAQ:ANIP), GlaxoSmithKline (NYSE:GSK), Oxford BioMedica (OTC:OXBDF), Transgene, Urigen Pharmaceuticals Inc. (OTC:URGP), and Vical, Inc. (NASDAQ:VICL).
Big pharma has renewed its interest in gene therapy.
In October 2010, GlaxoSmithKline (Glaxo), Fondazione Telethon and Fondazione San Raffaele announced a new strategic alliance to research and develop novel treatments to address rare genetic disorders, using gene therapy carried out on stem cells taken from the patient's bone marrow (ex vivo). The alliance capitalizes on research performed at the San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), a joint venture between Fondazione Telethon and Fondazione San Raffaele established in 1995.
Under the terms of the agreement, Glaxo gained an exclusive license to develop and commercialize an investigational gene therapy, for ADA Severe Combined Immune Deficiency (ADA-SCID), a rare and life-threatening immune deficiency, which affects approximately 350 children worldwide. Phase 1/Phase 2 studies have demonstrated the potential of this treatment option to restore long-term immune function and protect against severe infections in children with ADA deficiency.
Glaxo is co-developing with Fondazione Telethon and Fondazione San Raffaele six other applications of ex vivo stem cell therapy using a new gene transfer technology developed by HSR-TIGET scientists with the potential to treat a range of rare disorders, including metachromatic leukodystrophy and Wiskott-Aldrich Syndrome, beta-thalassemia, mucopolysaccharoidosis type I, globoid leukodystrophy, and chronic granulomatous disorder. .
Fondazione Telethon received an upfront 10 million euro payment from Glaxo and is eligible to receive further payments upon successful completion of a number of predetermined development milestones.
In September 2011, Merck & Co (NYSE:MRK) announced that it issued an exclusive license to FKD Therapies Oy, a recently formed company in Finland specializing in advanced gene based medicines to develop and commercialize its gene therapy portfolio.
On August 26, 2013, Sangamo BioSciences, Inc. (NASDAQ:SGMO) announced that it signed a definitive agreement to acquire Ceregene, Inc., a privately held biotechnology company focused on developing adeno-associated virus gene therapies.
On May 1, 2013, Oxford BioMedica, a British company, announced that it agreed to produce clinical trial material using its LentiVector gene delivery technology for Novartis's (NYSE:NVS) use as part of its CTL019 leukemia program, a collaboration with scientists at the University of Pennsylvania.
Through this production agreement, Oxford BioMedica will be responsible for manufacturing a lentiviral vector-encoding CTL019 technology, which Novartis et al. will use to transduce patients' T-cells ex vivo before they are reinfused. In its announcement, Oxford BioMedica noted that CTL019 targets the protein CD19, which is associated with a number of B-cell malignancies, including chronic lymphocytic leukemia, B-cell acute lymphocytic leukemia, and diffuse large B-cell lymphoma.
Under the terms of the agreement, Oxford BioMedica stands to make $3.9 million to $6.2 million during the first year of collaboration.
The US National Institutes of Health (NIH) clinicaltrials.gov website lists over 3,000 gene therapy trials, of which 1,300 are listed as recruiting. The vast majority of these are being run by academic investigators.
The excitement surrounding gene therapy was tempered on August 12, 2013 when shares of Vical plunged after the company announced that it was terminating its Allovectin (velimogene aliplasmid) program and focusing its resources on its infectious disease vaccine program. In a Phase 3 trial of patients with metastatic melanoma, Allovectin failed to demonstrate a statistically significant improvement versus first-line chemotherapy for either the primary endpoint of objective response rate at 24 weeks or more after randomization or the secondary endpoint of overall survival.

Lenti-D

bluebird's most advanced product candidate is Lenti-D, which the company is developing initially to treat patients with childhood cerebral adrenoleukodystrophy (CCALD).
CCALD is a rare, hereditary neurological disorder affecting young boys that is often fatal. CCALD is caused by mutations in the ABCD1 gene. bluebird bio scientists believe the disease can be treated with the ex vivo insertion of a functional copy of the ABCD1 gene into a patient's own HSCs to correct the aberrant expression of ALDP in patients with CCALD. HSCs derived from the patient's own body are called autologous HSCs. bluebird bio has named the autologous HSCs that have been modified to carry the functional copy of the ABCD1 gene as the final Lenti-D drug candidate.
bluebird bio performed a non-interventional retrospective data collection study. Comprised of 136 CCALD patients, the study assessed the course of the disease in patients who were left untreated and patients who received allogeneic HSCT. Researchers conducted a non-interventional retrospective data collection study that examined the historical clinical records from patients in the study to assess the typical course of the condition and the efficacy and safety of treatment options. The company believe the results of this study supported its approach of using autologous, gene-modified HSCs to treat CCALD, especially in light of several significant safety concerns commonly associated with the current standard of care, allogeneic HSCTs. Allogeneic HSCTs are the HSCTs from donors other than the patient.
The company believes that the results from a Phase 1/2 study in four patients with CCALD conducted by the company's scientific collaborators in France with an earlier generation lentiviral vector were useful in the design of its own trials to evaluate the efficacy and safety of Lenti-D.
Early clinical proof-of-concept results for two patients in a French study sponsored by the Institut National de la Santé et de la Recherche Médicale (National Institute of Health and Medical Research) (INSERM) produced promising results that were published in the November 2009 issue of Science.
In this pilot study, blood stem cells were removed from patients and genetically modified in the laboratory using a lentiviral vector to introduce a working copy of the ALD gene into the cells. The patients' modified cells were then infused back into the patients after they received a treatment to allow engraftment of the corrected cells. Initial results from this study demonstrated disease stabilization and no further disease progression in the two patients monitored for two years.
On May 20, 2010, Nathalie Cartier-Lacave, MD, study investigator and a director of research at the National Institute of Health and Medical Research at INSERM, presented additional data from the pilot study. Based on the continued follow-up of the first two patients as well as 20-month data on a third patient, the new data found that at three years, the two patients' functional ALD proteins remained stably expressed at 10% to 15% of circulating monocytes. Both continued to show neurological stabilization with no adverse effects noted. In the third patient, functional ALD protein remained stably expressed at 20 months. The new findings also demonstrated that the third patient's cognitive functioning was within normal range at 16 months. Although these results appear to be extremely promising, the researchers warned that the current data was insufficient to determine that the disease was stabilized. The researchers reported that the safety and therapeutic benefits of this procedure have been maintained for more than five years, and two additional patients have subsequently been treated.
Based on these promising early clinical proof of concept results, bluebird bio plans to initiate a larger Phase 2/3 clinical trial in CCALD in the United States and Europe in 2013 to evaluate Lenti-D's safety and efficacy in subjects with CCALD. The estimated completion date of the study (final data collection date for primary outcome measure) is August 2018.
In this clinical trial, up to 15 patients will be enrolled in order to obtain at least 12 subjects who will be followed over a 24-month period to assess the onset of major functional disabilities (MFDs), and other key assessments of disease progression. bluebird bio expects to initiate this study in the United States in late 2013. If successful, the company believes the results of this study could support submission of a Biologics License Application (BLA) and a Marketing Authorization Application (MAA) filing for Lenti-D.
On June 19, 2012, bluebird announced that both the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) granted an orphan drug designation to its investigational gene therapy product for the treatment of ALD.
"Receiving orphan drug designation is a positive step forward in our efforts to bring hope to ALD patients and their families," said David Davidson, M.D., chief medical officer of bluebird bio. "We believe our lentiviral technology has the potential to be a one-time transformative therapy for patients suffering from rare genetic disorders like ALD for whom there are limited treatment options. bluebird is committed to advancing the clinical and commercial development of our gene therapy platform because of the dramatic benefit it may have on the lives of patients."
Orphan drug designation, which is intended to facilitate drug development for rare diseases, provides benefits to the sponsor, including the potential for funding for certain clinical studies, study-design assistance, and several years of market exclusivity for the product upon regulatory approval.
In April 2013, the FDA informed bluebird bio that the Investigational New Drug application (IND) the company filed in March 2013 for a Phase 2/Phase 3 clinical study to evaluate Lenti-D's effect in preserving neurological function and stabilizing cerebral demyelination in subjects with CCALD was active.
Adrenoleukodystrophy (ALD) is a rare X-linked, inherited neurological disorder. In its most severe form, ALD causes a breakdown of the myelin sheath in the brain (a protective and insulating membrane that surrounds nerve cells) and progressive dysfunction of the adrenal glands. This damage can result in decreased motor coordination and function, visual and hearing disturbances, the loss of cognitive function, dementia, seizures, adrenal dysfunction and other complications, including death.
Also known as "Lorenzo's Oil disease," named after the 1993 film starring Susan Sarandon and Nick Nolte, ALD is estimated to affect approximately one in every 20,000 boys worldwide. ALD, is identified in about 120 young boys in the United States each year. In the childhood cerebral form (CCALD), symptoms usually occur between the ages of 3 and 15 years. Symptoms often progress rapidly and can lead to a vegetative state and death.
Currently, the only effective treatment option is allogeneic hematopoietic stem cell transplant (HSCT), in which the patient is treated with HSCs containing the properly functioning copy of the gene, which is contributed by a donor other than the patient. Unfortunately, allogenic HSCT has significant morbidity and mortality, particularly in patients who undergo non-sibling matched allogeneic HSCT. Sibling matched donors are only typically available for less than 30% of patients. Yet even in these patients, severe graft versus host disease (GVHD) is the lead cause of mortality after transplantation. It is estimated that even at the best transplant centers, approximately 15% of patients suffer transplant related mortality, and approximately 30% experience GVHD.
bluebird bio is developing Lenti‐D, a lentivirus based autologous cell therapy, as a safer alternative to allogeneic HSCT. bluebird bio scientists have designed Lenti-D to restore functional ALDP production in a patient's own hematopoietic stem cells, eliminating the risk of GVHD.

LentiGlobin

bluebird bio is also developing LentiGlobin to treat patients with ß-thalassemia and sickle cell disease (SCD).
ß-thalassemia is a rare hereditary blood disorder caused by a genetic abnormality of the ß-globin gene resulting in defective red blood cells. Symptoms of ß-thalassemia include severe anemia, splenomegaly, marrow expansion, bone deformities and iron overload in major organs. According to Thalassemia International Federation, about 288,000 patients with ß-thalassemia major are alive and registered as receiving regular treatment around the world, of which it is estimated that about 15,000 live in the United States and Europe.
Patients with beta-thalassemia major (the most severe form of beta-thalassemia) receive chronic blood transfusions aimed at maintaining a steady hemoglobin levels to treat their severe anemia. Chronic blood transfusions can be effective at preventing many symptoms of childhood beta thalassemia major, but chronic transfusions introduce a large iron overload, which over time may lead to mortality through iron-associated heart and liver toxicity. In order to prevent iron overload-associated risks, patients must adhere to daily iron chelation regimens. Poor compliance with chelation regimens remains a key challenge, and even with transfusion and iron chelation therapies, overall survival is significantly reduced.
The only potentially curative therapy for beta-thalassemia is allogeneic hematopoietic stem cell transplant (HSCT). Due to the significant risk of transplant related morbidity and mortality, transplants are offered primarily only to pediatric patients with matched sibling donors, which occurs in less than 25% of all cases. Allogeneic HSCT carries a significant risk of morbidity and mortality related to serious infection, graft failure and graft-versus-host-disease.
SCD is a hereditary blood disorder resulting from a mutation in the ß-globin gene that causes polymerization of hemoglobin proteins and abnormal red blood cell function. SCD is characterized by anemia, vaso-occlusive crisis (a common complication of SCD in which there is severe pain due to obstructed blood flow in the bones, joints, lungs, liver, spleen, kidney, eye, or central nervous system), infections, stroke, overall poor quality of life and early death in a large subset of patients. The global incidence of SCD is estimated to be 250,000 to 300,000 births annually, and the global prevalence of the disease is estimated to be about 20 million to 25 million.
According to GBI Research, the SCD market will be the fastest growing of the three markets during the forecast period with a Compound Annual Growth Rate (OTCPK:CAGR) of 9% seeing it reach $70 million in 2019. The thalassemia market will grow at a lower CAGR of 7% to reach $59 million in 2019.
bluebird's approach involves the insertion of a single codon variant of the normal ß-globin gene, referred to as T87Q, into the patient's own HSCs via an HIV-1 based lentiviral vector to restore expression of the ß-globin protein required for hemoglobin production. The codon variant is also used as a biomarker to quantify expression levels of ß-globin protein derived from the vector, and provides strong anti-sickling properties in the context of SCD. bluebird bio refers to the gene-modified HSCs as the final LentiGlobin drug product.
On September 15, 2010, bluebird bio announced promising Phase 1/Phase 2 data highlighting positive results of LentiGlobin gene therapy treatment in a young adult with severe beta-thalassemia that was published in the September 2010 issue of Nature,
The patient, who had been transfusion dependent since early childhood, became transfusion independent for the past 21 months, more than two years after treatment with the LentiGlobin vector. The study also identified a subset of cells with the corrected beta-globin gene that overexpressed a truncated form of a gene called HMGA2. The patient has not experienced any adverse events. The data show that while early on, the HMGA2 clone was a significant portion of the corrected cells, the clone levels had declined at the time the paper was prepared, and further follow up indicates the decline is continuing.
"Although based on the first treated patient, we believe these results are impressive and illustrate for the first time the significant potential for treatment of beta hemoglobinopathies using lentiviral beta-globin gene transfer in the context of autologous stem cell transplant," said Philippe Leboulch, MD, senior author of the study and head of the Institute of Emerging Diseases and Innovative Therapies of CEA and INSERM; professor of medicine, University of Paris and visiting professor, Harvard Medical School. "For beta-thalassemia, we have worked intensely for almost 20 years to design, develop and manufacture LentiGlobin to provide a sustained high level hemoglobin production, resulting in a major clinical benefit. It has been very rewarding to follow this patient as his life has dramatically improved since receiving our treatment."

"For the first time, a patient with severe beta-thalassemia is living without the need for transfusions over a sustained period of time," said Marina Cavazzana-Calvo, MD, first co-author of the study and professor of hematology, University of Paris and chief of Cell and Gene Therapy Department, Necker-Enfants Malades Hospital in Paris. Salima Hacein-Bey-Abina, PhD, professor of immunology, University of Paris, added, "These results are not only important due to the tremendous medical need that exists for thalassemia patients around the world, but also represents a significant step forward for the field of autologous stem cell therapy as an emerging therapeutic modality."
On March 16, 2011, bluebird bio announced that it had entered into an agreement with the French Muscular Dystrophy Association (AFM), a French non-profit entity, whereby the company will receive an initial amount of approximately $1.4 million in cash to support the development of LentiGlobin. As part of the research agreement, bluebird bio has the option to draw upon an additional amount of up to $2.8 million in credit toward the manufacturing of cGMP clinical trial material at Généthon. In December 2010, bluebird bio entered into an agreement with Généthon designed to enable substantial advances in the existing manufacturing process of lentiviral vectors for the benefit of both partners.
In a Phase 1/Phase 2 study of patients with ß-thalassemia major being conducted by the company's scientific collaborators in France with an earlier generation of its LentiGlobin vector called HPV569, data have provided initial evidence of transfusion independence following treatment with gene modified HSCs. Now bluebird bio plans to use its new LentiGlobin vector for studies based on higher transduction efficiency and expression of ß-globin protein in target cells as compared to the HPV569 vector. bluebird bio initiated a study in France using a revised clinical protocol based on the use of LentiGlobin instead of HPV569. This Phase 1/Phase 2 continuation study, known as the HGB-205 Study, will enroll up to seven additional subjects with ß-thalassemia major or SCD to evaluate transfusion requirements post-transplant, as well as the number of hospitalization days post-transplant discharge. In SCD patients only, efficacy will also be measured based on the number of vaso-occlusive crises or acute chest syndrome events
The company also plans to initiate a Phase 1/Phase 2 clinical study in the United States to evaluate its LentiGlobin product candidate in increasing hemoglobin production and eliminating or reducing transfusion dependence in patients with ß-thalassemia major, which the company refers to as the HGB-204 Study. Up to 15 adults will be enrolled to evaluate production of hemoglobin containing ßA-T87Q-globin for the six-month period between 18 and 24 months post-transplant, followed by long-term monitoring to assess safety and efficacy beyond the initial 24 months. bluebird bio expects to submit an IND with the FDA in 2014 to evaluate LentiGlobin in patients with SCD
In October 2012, bluebird bio announced that the California Institute for Regenerative Medicine (CIRM) approved an award to the company for $9.3 million for the first round of its new Strategic Partnership Awards initiative. The award is to support a Phase 1/2 study to evaluate the safety and efficacy of LentiGlobin for the treatment of beta-thalassemia.

CAR T

On March 21, 2013, bluebird announced the formation of a broad, global strategic collaboration with Celgene to discover, develop and commercialize novel disease- altering gene therapies in oncology.
CAR T-cell therapy represents a promising, emerging approach to treating cancer. Blood is withdrawn from a patient and the T-cells are then extracted from a patient's blood. These cells are then genetically modified to recognize and attack cancer cells and then re-introduced into the patient's blood. The patient's genetically modified cells are intended to bind to and kill the target cancer cells.
Celgene has also entered into a separate strategic collaboration in the CAR T-cell field with the Center for Cell and Gene Therapy at Baylor College of Medicine, Texas Children's Hospital and The Methodist Hospital, Houston, led by Malcolm Brenner, M.D., Ph.D., professor, Department of Molecular and Human Genetics and the director, Center for Cell and Gene Therapy. bluebird bio, Celgene and Dr. Brenner's team will work collaboratively to advance and develop existing and new products and programs in the CAR T-cell field.
The collaboration is applying gene therapy technology to genetically modify a patient's own T-cells, known as chimeric antigen receptor (CAR) T-cells, to target and destroy cancer cells. The multi-year research and development collaboration has the potential to lead to the development and commercialization of multiple CAR T-cell products.
Under the agreement, Celgene has an option to license any products resulting from the collaboration after the completion of a Phase 1 clinical study for each such product. bluebird is responsible for research and development activity through Phase 1 studies.
"The genetic manipulation of autologous T-cells is a new frontier in oncology, one that shows early promise in emerging clinical trials," said Tom Daniel, president, research & early development at Celgene. "We see strong prospects for this collaboration between Celgene, bluebird bio and Baylor College of Medicine's experienced leaders in this emerging field, led by Dr. Brenner, to advance this innovative approach to intractable problems in oncology."
Financial terms of the agreement include an upfront payment and up to $225 million per production potential option fees and clinical and regulatory milestones. bluebird also has the right to participate in the development and commercialization of any licensed products resulting from the collaboration through a 50/50 co-development and profit share in the United States in exchange for a reduction of milestones. Royalties would also be paid in regions where there is no profit share including in the United States if bluebird bio declines to exercise their co-development and profit sharing rights.

Finances

On August. 14, 2013, bluebird bio reported financial results and operational highlights for the quarter ended June 30, 2013.
Revenues were $6.3 million during the quarter ended June 30, 2013 compared to $0.1 million for the quarter ended June 30, 2012 and include amounts allocated to research and development services from the Celgene collaboration.
Net cash provided by operating activities during the six months ended June 30, 2013 was $57.8 million. bluebird bio held $228.8 million in cash and cash equivalents as of June 30, 2013.
Total operating expenses for the quarter ended June 30, 2013 were $10.5 million as compared to $4.7 million for the same period in 2012.
bluebird bio reported a net loss of $4.6 million, or $2.13 per share, for the quarter ended June 30, 2013, as compared to a net loss of $4.6 million, or $23.21 per share, for the quarter ended June 30, 2012. The net loss applicable to basic and diluted common shareholders was $4.6 million for the quarter ended June 30, 2013, as compared to $5.8 million for the quarter ended June 30, 2012.

Conclusion

Most analysts are positive about bluebird bio. Motley Fool readers gave the stock a three star rating. Zacks has a 3 or Hold rating on the stock.
On July 30, 2013, Cowen and Company initiated coverage of bluebird bio with an Outperform rating and a $44 price target.
On July 15, 2013, both Wedbush and JPMorgan initiated coverage of bluebird bio with Outperform ratings. Wedbush put a $32.25 price target on the stock. Both Bank of America and Canaccord Genuity began coverage of bluebird bio with a Buy rating. Canaccord set a $45 price target, and Bank of America put a $42 price target on the stock. JPMorgan gave bluebird bio a $44 price target.
Top institutional holders of bluebird bio stock are Ameriprise Financial Inc., Camber Capital Management, LLC, and Citadel Advisors LLC. Top mutual fund holders are Fidelity Contrafund, Prudential Jennison Health Sciences Fund, American Funds Insurance Ser-Global Small Capitalization Fund, Fidelity Growth Company Fund, Fidelity Growth Company Fund, Waddell & Reed Advisors Funds-Science & Technology Fund, Ivy Science & Technology Fund, Fidelity Advisor New Insights Fund, Fidelity Blue Chip Growth Fund, and Franklin Strategic Series-Franklin Biotechnology Discovery Fund.
Nick Leschly has served as the company's president and chief executive officer since September 2010. Previously, he served as the company's interim chief executive officer from March 2010 to September 2010. Formerly a partner of Third Rock Ventures, L.P. since its founding in 2007, Leschly played an integral role in the overall formation, development and business strategy of several of Third Rock's portfolio companies, including Agios Pharmaceuticals, Inc. (NASDAQ:AGIO) and Edimer Pharmaceuticals, Inc.
bluebird bio's gene therapy platform holds the promise of delivering a one-time treatment for a wide variety of diseases. The company's initial focus is on genetic diseases caused by a known mutation in a single gene. Through the application of gene therapy, bluebird bio is developing product candidates designed to insert a functional gene into a patient, potentially correcting their underlying disease. In addition to genetic diseases, bluebird bio and its partners are also utilizing this technology to develop product candidates intended to treat various cancers, by targeting a patient's own immune system to seek out and kill cancer cells in the body, offering a new approach to cancer treatment.
Although gene therapy shows great promise for a number of currently incurable diseases, the technique remains risky and is still under study to ensure that it is safe and effective. Currently, gene therapy is only being tested for the treatment of diseases that have no other cures.
bluebird bio is well positioned to drive the continued advancement of gene therapy technology for the treatment of severe genetic and orphan diseases. The company has assembled extensive expertise in viral vector design, manufacturing and gene transfer, a broad intellectual property estate, an experienced management team and a high caliber group of scientific advisors and key opinion leaders.
Disclosure: I am long CELG, MRK. I wrote this article myself, and it expresses my own opinions. I am not receiving compensation for it (other than from Seeking Alpha). I have no business relationship with any company whose stock is mentioned in this article

No comments:

Post a Comment