From Challenge to Opportunity: How Shwachman–Diamond Syndrome Became a Promising Target for Therapy Development

IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY
Eszter S. Hars, Lisa J. McReynolds
{"title":"From Challenge to Opportunity: How Shwachman–Diamond Syndrome Became a Promising Target for Therapy Development","authors":"Eszter S. Hars,&nbsp;Lisa J. McReynolds","doi":"10.1002/cpt.3393","DOIUrl":null,"url":null,"abstract":"<p>\n <b>Rare diseases affect over 30 million people in the United States and 300 million globally, yet 95% lack FDA-approved treatments. Rare disease therapy development poses unique challenges and opportunities. Shwachman–Diamond syndrome (SDS) is emerging as a model rare disease due to its uniform genetics, robust molecular understanding, and mature research infrastructure—including de-risking through model development, regulatory engagement, patient community development, ICD-10 implementation, and data access, driven by the SDS Alliance—a research-focused patient advocacy organization.</b>\n </p><p>Rare disease connects us all. Every one of us knows someone impacted by one of over 10,000 rare diseases, which combined affect 300 million people globally and over 30 million people in the US—nearly 1 in 10. 95% of rare diseases have no FDA-approved treatments, but the tides are changing. Drugs that target rare diseases represent over half of all novel drugs and biologics approved by the FDA in recent years. This did not happen overnight or by chance. The Orphan Drug Act of 1983 and the relentless advocacy of leading rare disease organizations made this possible.<span><sup>1</sup></span></p><p>Developing drugs for rare diseases poses unique challenges and opportunities. Beyond financial considerations, developers face hurdles such as recruiting enough patients for clinical trials, obtaining natural history data, and designing clinical trials that can convincingly demonstrate safety and efficacy to regulators and patients. Opportunities include new niche markets with substantial financial potential and the intangible rewards of bringing therapies to patients in desperate need, witnessing firsthand the life-changing positive impact on families and communities.</p><p>Given the large number of distinct rare diseases and the high risks involved in therapy development, what makes a rare disease a compelling target for investment? The more that is known about the disease mechanism, the more that patients are connected, and the more that relevant research tools and infrastructure are developed, the better. But often it takes serendipity and time. Small molecule drugs developed for a common condition may be a great fit for a rare disease (drug repurposing) based on its mechanism of action. Modern gene editing tools open doors for addressing the underlying cause of rare genetic disorders. Precision medicine is the new frontier, offering hope not only to rare disease patients but to all of us.</p><p>SDS is a rare genetic disorder that affects about 1:100,000 births, and thousands of people globally, many undiagnosed due to variable presentation of symptoms.<span><sup>2, 3</sup></span> It impacts multiple organ systems and significantly increases the risk of leukemia, with poor outcomes.<span><sup>4, 5</sup></span> By age 30, about 30% of SDS patients develop acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), or severe bone marrow failure, often necessitating a hematopoietic cell transplant (HCT)—a high-risk procedure with uncertain long-term effects. The outcomes for AML in SDS patients are particularly grim.<span><sup>4</sup></span></p><p>Other common symptoms include failure to thrive, pain, and malnutrition due to exocrine pancreatic insufficiency (EPI), elevated liver enzymes, respiratory issues, skeletal abnormalities, and cognitive delays.<span><sup>2, 3</sup></span> The severity of symptoms varies widely from patient to patient, causing frequently missed or delayed diagnoses, and putting patients at risk for unnecessary suffering and life-threatening complications.</p><p>Most cases of SDS are caused by pathogenic variants in the essential gene <i>SBDS</i>,<span><sup>6</sup></span> typically inherited from unsuspecting carrier parents in an autosomal recessive pattern, or caused <i>de-novo</i> by gene conversion/recombination with an adjacent pseudogene, <i>SBDSP1</i>. The SBDS protein plays a critical role in ribosome biogenesis by catalyzing the displacement of eIF6 from the large ribosomal subunit, thereby allowing the large and small ribosomal subunits to join and form a working ribosome.<span><sup>6</sup></span> Ribosomes are large protein complexes that translate the genetic information encoded in mRNA into proteins, playing a key role in the central dogma of molecular biology and being fundamental to life. In SDS, there are not enough working ribosomes to meet the protein demand of cells. Recent publications suggest that the highly increased risk of developing leukemia is due to “reduced fitness” of hematopoietic stem cells (HSCs), causing a selection pressure favoring cells that acquire maladaptive somatic mutations, such as TP53.<span><sup>6, 7</sup></span></p><p>Currently, there is no targeted therapy for SDS. Patients are left anxious about possible life-threatening complications and invasive surveillance to look for concerning findings to trigger HCT. On the positive side, day-to-day quality of life has significantly improved thanks to advances in symptomatic treatments, such as pancreatic enzyme replacement therapy (PERT) to manage the effects of EPI, granulocyte-colony stimulating factor (G-CSF) to improve neutropenia, and others.</p><p>There is a wide range of pre-clinical research efforts aimed at addressing the underlying cause of SDS. Several programs are exploring base- and prime editing to fix the genetic cause; there are small molecule screening and development efforts to de-stabilize the binding of eIF6 to overcome the SBDS defect, based on learning from somatic genetic rescue<span><sup>8</sup></span>; antisense oligonucleotides (ASOs) have been shown to increase wild-type SBDS expression; plus, additional programs are looking at targeting other common SBDS pathogenic variants with nonsense suppressor drugs.<span><sup>9</sup></span></p><p>SDS has several characteristics that make it an ideal model rare disease and target for therapy development (see <b>Figure</b> 1).</p><p>First, the genetics are well understood and extremely uniform. Over 90% of patients not only have pathogenic variants in the same gene (<i>SBDS</i>) but also have at least one copy of a specific splice-site mutation (known as c.258+2T&gt;C). This common variant allows a small amount of wild-type protein to be made, making SDS compatible with life, and making it a good candidate for several therapeutic modalities.<span><sup>10</sup></span></p><p>Second, the primary organ system of concern, the bone marrow and in particular the HSCs, are readily accessible by a variety of therapeutic modalities, including small molecules and gene therapy (i.e., base and prime editing)—both <i>ex vivo</i> (via HCT as in severe combined immunodeficiency (SCID) and sickle cell disease) and <i>in vivo</i> (a rapidly evolving field).</p><p>Third, due to the long history of SDS research, several natural history studies have been collecting clinical data for years or decades and are ready to support clinical development. Additional data resources are available as well, as discussed below.</p><p>To further de-risk SDS therapy development, the Shwachman-Diamond Syndrome Alliance—a global research-focused patient advocacy nonprofit organization—has launched several programs to develop critical research tools and infrastructure. These programs fall into four broad categories: model systems, regulatory engagement, patient community development, and data access (see <b>Figure</b> 1).</p><p>Reflecting on the SDS Alliance's experience with positioning SDS as a model rare disease and emulating the strategies of successful rare disease organizations in recent years, it is evident that modest investments in strategic tools and infrastructure can significantly de-risk rare diseases and incentivize new therapeutic development and drug repurposing. We hope that other rare disease communities will benefit from these approaches and that drug developers continue to recognize the value of partnering with patient communities to pursue therapies and cures for the thousands of rare diseases affecting millions of people. Shwachman–Diamond syndrome is ready.</p><p>This work was supported by The Chan Zuckerberg Initiative, a research grant from the University of Pennsylvania Orphan Disease Center in partnership with the Shwachman-Diamond Syndrome Alliance, and the SDS patient community. LJM was funded by the Intramural Research Program of the National Cancer Institute.</p><p>The authors declare no competing interests for this work.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"116 6","pages":"1377-1380"},"PeriodicalIF":6.3000,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11567794/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacology & Therapeutics","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cpt.3393","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

Abstract

Rare diseases affect over 30 million people in the United States and 300 million globally, yet 95% lack FDA-approved treatments. Rare disease therapy development poses unique challenges and opportunities. Shwachman–Diamond syndrome (SDS) is emerging as a model rare disease due to its uniform genetics, robust molecular understanding, and mature research infrastructure—including de-risking through model development, regulatory engagement, patient community development, ICD-10 implementation, and data access, driven by the SDS Alliance—a research-focused patient advocacy organization.

Rare disease connects us all. Every one of us knows someone impacted by one of over 10,000 rare diseases, which combined affect 300 million people globally and over 30 million people in the US—nearly 1 in 10. 95% of rare diseases have no FDA-approved treatments, but the tides are changing. Drugs that target rare diseases represent over half of all novel drugs and biologics approved by the FDA in recent years. This did not happen overnight or by chance. The Orphan Drug Act of 1983 and the relentless advocacy of leading rare disease organizations made this possible.1

Developing drugs for rare diseases poses unique challenges and opportunities. Beyond financial considerations, developers face hurdles such as recruiting enough patients for clinical trials, obtaining natural history data, and designing clinical trials that can convincingly demonstrate safety and efficacy to regulators and patients. Opportunities include new niche markets with substantial financial potential and the intangible rewards of bringing therapies to patients in desperate need, witnessing firsthand the life-changing positive impact on families and communities.

Given the large number of distinct rare diseases and the high risks involved in therapy development, what makes a rare disease a compelling target for investment? The more that is known about the disease mechanism, the more that patients are connected, and the more that relevant research tools and infrastructure are developed, the better. But often it takes serendipity and time. Small molecule drugs developed for a common condition may be a great fit for a rare disease (drug repurposing) based on its mechanism of action. Modern gene editing tools open doors for addressing the underlying cause of rare genetic disorders. Precision medicine is the new frontier, offering hope not only to rare disease patients but to all of us.

SDS is a rare genetic disorder that affects about 1:100,000 births, and thousands of people globally, many undiagnosed due to variable presentation of symptoms.2, 3 It impacts multiple organ systems and significantly increases the risk of leukemia, with poor outcomes.4, 5 By age 30, about 30% of SDS patients develop acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), or severe bone marrow failure, often necessitating a hematopoietic cell transplant (HCT)—a high-risk procedure with uncertain long-term effects. The outcomes for AML in SDS patients are particularly grim.4

Other common symptoms include failure to thrive, pain, and malnutrition due to exocrine pancreatic insufficiency (EPI), elevated liver enzymes, respiratory issues, skeletal abnormalities, and cognitive delays.2, 3 The severity of symptoms varies widely from patient to patient, causing frequently missed or delayed diagnoses, and putting patients at risk for unnecessary suffering and life-threatening complications.

Most cases of SDS are caused by pathogenic variants in the essential gene SBDS,6 typically inherited from unsuspecting carrier parents in an autosomal recessive pattern, or caused de-novo by gene conversion/recombination with an adjacent pseudogene, SBDSP1. The SBDS protein plays a critical role in ribosome biogenesis by catalyzing the displacement of eIF6 from the large ribosomal subunit, thereby allowing the large and small ribosomal subunits to join and form a working ribosome.6 Ribosomes are large protein complexes that translate the genetic information encoded in mRNA into proteins, playing a key role in the central dogma of molecular biology and being fundamental to life. In SDS, there are not enough working ribosomes to meet the protein demand of cells. Recent publications suggest that the highly increased risk of developing leukemia is due to “reduced fitness” of hematopoietic stem cells (HSCs), causing a selection pressure favoring cells that acquire maladaptive somatic mutations, such as TP53.6, 7

Currently, there is no targeted therapy for SDS. Patients are left anxious about possible life-threatening complications and invasive surveillance to look for concerning findings to trigger HCT. On the positive side, day-to-day quality of life has significantly improved thanks to advances in symptomatic treatments, such as pancreatic enzyme replacement therapy (PERT) to manage the effects of EPI, granulocyte-colony stimulating factor (G-CSF) to improve neutropenia, and others.

There is a wide range of pre-clinical research efforts aimed at addressing the underlying cause of SDS. Several programs are exploring base- and prime editing to fix the genetic cause; there are small molecule screening and development efforts to de-stabilize the binding of eIF6 to overcome the SBDS defect, based on learning from somatic genetic rescue8; antisense oligonucleotides (ASOs) have been shown to increase wild-type SBDS expression; plus, additional programs are looking at targeting other common SBDS pathogenic variants with nonsense suppressor drugs.9

SDS has several characteristics that make it an ideal model rare disease and target for therapy development (see Figure 1).

First, the genetics are well understood and extremely uniform. Over 90% of patients not only have pathogenic variants in the same gene (SBDS) but also have at least one copy of a specific splice-site mutation (known as c.258+2T>C). This common variant allows a small amount of wild-type protein to be made, making SDS compatible with life, and making it a good candidate for several therapeutic modalities.10

Second, the primary organ system of concern, the bone marrow and in particular the HSCs, are readily accessible by a variety of therapeutic modalities, including small molecules and gene therapy (i.e., base and prime editing)—both ex vivo (via HCT as in severe combined immunodeficiency (SCID) and sickle cell disease) and in vivo (a rapidly evolving field).

Third, due to the long history of SDS research, several natural history studies have been collecting clinical data for years or decades and are ready to support clinical development. Additional data resources are available as well, as discussed below.

To further de-risk SDS therapy development, the Shwachman-Diamond Syndrome Alliance—a global research-focused patient advocacy nonprofit organization—has launched several programs to develop critical research tools and infrastructure. These programs fall into four broad categories: model systems, regulatory engagement, patient community development, and data access (see Figure 1).

Reflecting on the SDS Alliance's experience with positioning SDS as a model rare disease and emulating the strategies of successful rare disease organizations in recent years, it is evident that modest investments in strategic tools and infrastructure can significantly de-risk rare diseases and incentivize new therapeutic development and drug repurposing. We hope that other rare disease communities will benefit from these approaches and that drug developers continue to recognize the value of partnering with patient communities to pursue therapies and cures for the thousands of rare diseases affecting millions of people. Shwachman–Diamond syndrome is ready.

This work was supported by The Chan Zuckerberg Initiative, a research grant from the University of Pennsylvania Orphan Disease Center in partnership with the Shwachman-Diamond Syndrome Alliance, and the SDS patient community. LJM was funded by the Intramural Research Program of the National Cancer Institute.

The authors declare no competing interests for this work.

Abstract Image

从挑战到机遇:舒瓦赫曼-钻石综合征如何成为有望开发的治疗目标?
临床前研究工作范围广泛,旨在解决 SBDS 的根本原因。有几个项目正在探索通过碱基和质粒编辑来修复遗传病因;有一些小分子筛选和开发项目,根据体细胞遗传救援8 的经验,努力去稳定 eIF6 的结合,以克服 SBDS 缺陷;反义寡核苷酸(ASO)已被证明可以增加野生型 SBDS 的表达;此外,还有一些项目正在研究针对其他常见 SBDS 致病变体的无义抑制药物。SBDS 有几个特点,使其成为理想的罕见病模型和治疗开发靶点(见图 1)。超过 90% 的患者不仅在同一基因中存在致病变异(SBDS),而且至少有一个特定剪接位点突变的拷贝(称为 c.258+2T&gt;C)。这种常见变异允许制造少量野生型蛋白质,使 SDS 与生命相容,并使其成为几种治疗方法的良好候选者。10 其次,主要的相关器官系统,即骨髓,尤其是造血干细胞,可随时通过各种治疗方法获得,包括小分子和基因治疗(即碱基和质粒编辑)、第三,由于 SDS 研究历史悠久,一些自然史研究已收集了数年或数十年的临床数据,可随时为临床开发提供支持。为了进一步降低 SDS 治疗开发的风险,舒瓦赫曼-钻石综合征联盟(Shwachman-Diamond Syndrome Alliance)--一个以研究为中心的全球性非营利性患者权益组织--已经启动了几项计划来开发关键的研究工具和基础设施。这些计划分为四大类:模型系统、监管参与、患者社区发展和数据访问(见图 1)。反思 SDS 联盟近年来将 SDS 定位为罕见病模型以及效仿成功罕见病组织的策略的经验,我们可以明显看出,对战略工具和基础设施的适度投资可以大大降低罕见病的风险,并激励新疗法的开发和药物的再利用。我们希望其他罕见病社区也能从这些方法中受益,并希望药物开发商继续认识到与患者社区合作的价值,为影响数百万人的数千种罕见病寻求治疗方法和疗法。这项工作得到了陈-扎克伯格倡议(Chan Zuckerberg Initiative)、宾夕法尼亚大学孤儿病中心(University of Pennsylvania Orphan Disease Center)与舒瓦赫曼-钻石综合征联盟(Shwachman-Diamond Syndrome Alliance)以及舒瓦赫曼-钻石综合征患者社区的合作研究基金的支持。LJM得到了美国国家癌症研究所校内研究项目的资助。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
12.70
自引率
7.50%
发文量
290
审稿时长
2 months
期刊介绍: Clinical Pharmacology & Therapeutics (CPT) is the authoritative cross-disciplinary journal in experimental and clinical medicine devoted to publishing advances in the nature, action, efficacy, and evaluation of therapeutics. CPT welcomes original Articles in the emerging areas of translational, predictive and personalized medicine; new therapeutic modalities including gene and cell therapies; pharmacogenomics, proteomics and metabolomics; bioinformation and applied systems biology complementing areas of pharmacokinetics and pharmacodynamics, human investigation and clinical trials, pharmacovigilence, pharmacoepidemiology, pharmacometrics, and population pharmacology.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信