Tingting - Current State of Developing Advanced Therapies For Rare Diseases in The European Union

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Expert Opinion on Orphan Drugs

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ieod20

Current state of developing advanced therapies


for rare diseases in the European Union

Tingting Qiu , Yitong Wang , Monique Dabbous , Eve Hanna , Ru Han ,


Shuyao Liang & Mondher Toumi

To cite this article: Tingting Qiu , Yitong Wang , Monique Dabbous , Eve Hanna , Ru Han ,
Shuyao Liang & Mondher Toumi (2020) Current state of developing advanced therapies for
rare diseases in the European Union, Expert Opinion on Orphan Drugs, 8:10, 417-429, DOI:
10.1080/21678707.2020.1835640

To link to this article: https://doi.org/10.1080/21678707.2020.1835640

View supplementary material

Published online: 21 Oct 2020.

Submit your article to this journal

Article views: 445

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ieod20
EXPERT OPINION ON ORPHAN DRUGS
2020, VOL. 8, NO. 10, 417–429
https://doi.org/10.1080/21678707.2020.1835640

ORIGINAL RESEARCH

Current state of developing advanced therapies for rare diseases in the European
Union
Tingting Qiua, Yitong Wanga, Monique Dabbousa, Eve Hannab, Ru Hana, Shuyao Liang a
and Mondher Toumi a

a
Department of Public Health, Aix-Marseille University, Marseille, France; bDepartment of Price, Reimbursement and Market Access, Creativ-
ceutical, Paris, France

ABSTRACT ARTICLE HISTORY


Objective: Advanced Therapy Medicinal Products (ATMPs) present significant therapeutic advantages Received 25 June 2020
for inherited rare diseases. However, the development of orphan ATMPs is challenging due to their Accepted 8 October 2020
complexity and unpredictable biological activity. This study aims to comprehensively describe the
KEYWORDS
current state of orphan ATMPs in Europe.
Advanced therapy medicinal
Methods: Orphan drugs (ODs) granted by European Commission until March 2020 were investigated. products; european union;
The characteristic of diseases and ATMPs were extracted from the public summary reports of ODs from landscape; clinical trials;
Committee for Orphan Medicinal Products. The methodology for the pivotal studies of ATMPs was methodology
extracted.
Results: A total of 274 ATMPs were identified, covering 116 rare diseases, with metabolic, optical, and
oncologic diseases being the most targeted. A total of 158 ATMPs were indicated for life-threatening
diseases, 129 ATMPs targeted diseases currently lacking authorized or satisfactory treatment available.
Twenty-eight ATMPs are being investigated in the phase II/IIII or phase III studies. The median patient
size of pivotal studies was 127, 15 were open-label studies, 8 were single-arm trials, and 14 reported
surrogate outcomes.
Conclusion: There are rapid growths in developing ATMPs for life-threatening diseases with high
unmet clinical needs. Optimizing the study methodology and exploring innovative design to facilitate
the market access is paramount.

1. Introduction distinct regulatory programs for ATMPs have been established


in several countries in order to facilitate the MA of these
Rare diseases are increasingly considered a high public health
innovative therapies. For instance, the regenerative medicine
priority in many countries. Legislation for rare diseases has
advanced therapy (RMAT) designation has promoted exten­
been established in order to offset the potential losses of
sive interactions with the FDA beginning in very early stages
developing drugs in such a niche market, including non-
of drug development [8,9]. A steady market growth in the field
financial and financial incentives to pharmaceutical compa­
of ATMPs is being witnessed and the global ATMPs market is
nies, national rare disease initiatives, and expedited market
predicted to exceed 35.4 USD billion by 2026 [10].
authorization (MA) programs [1,2]. Such strategies have sig­
ODs have gained great attention from pharmaceutical
nificantly stimulated the research efforts, with more than 600
companies due to the supportive regulatory environment.
orphan drugs (ODs) approved worldwide [3] and the global
More importantly, the high prices of ODs, in order to recover
ODs market estimated to reach 209 USD billion by 2022 [4].
the development investment from small patient pool, also
The fast growth in the development of ODs is partly driven
have generate large revenue potential [11,12]. Compared to
by the improved knowledge of underlying genetic causes of
traditional ODs, ATMPs could result in extra costs driven by
rare diseases, given that approximately 80% of rare diseases
the personalized nature of the medicines, meaning that
are composed of genetic components [5]. The technological
some ATMPs cannot be prepared, tested, and manufactured
progress in advanced therapy medicinal products (ATMPs)
in bulk and, therefore, involve higher costs for clinical deliv­
represents great opportunity to treat rare and inherited dis­
ery [13]. Furthermore, ATMPs are associated with additional
eases, even for those previously perceived as incurable illness
value elements to those of ODs due to its curative potential
[6]. According to the European regulation EC No. 1394/2007
through the limited-time administration [14]. Undoubtedly,
[7], ATMPs were classified as gene therapy medicinal products
orphan ATMPs hold both pronounced importance for dis­
(GTMPs), somatic cell therapy medicinal products (sCTMPs),
ease treatment and enormous commercialization potential.
tissue-engineered products (TEPs) and combined products.
For example, nusinersen (Spinraza®), the first treatment
Like ODs, ATMPs addressing high unmet clinical needs possess
approved for spinal muscular atrophy (SMA), costs 750,000
a high potential for expedited MA pathways. Furthermore,
USD in the first year and 375,000 USD annually afterward.

CONTACT Tingting Qiu tingting.qiu@etu.univ-amu.fr Department of Public Health, Aix-Marseille University, Marseille, France
Supplemental data for this article can be accessed here.
© 2020 Informa UK Limited, trading as Taylor & Francis Group
418 T. QIU ET AL.

This placed Spinraza® amongst the priciest drugs in the The current development phases were defined as pre-clinical,
world post-market launch [15]. However, onasemnogene phase I, phase I/II, phase II/III, phase III, under evaluation, author­
abeparvovec-xioi (Zolgensma®), as the first gene therapy ized, rejected, and withdrawn from the EU market. Phase II/III
approved for SMA, was priced at 2.1 USD million per patient studies and phase III studies were regarded as late-stage devel­
for a single injection administration [16], making it the most opment. The study designs for ATMPs in phase II/III or phase III
expensive drug on the global market. studies were further investigated in relation to randomization
The purpose of this study was to conduct a comprehensive method, masking, comparator selection, report of outcomes,
analysis for the ATMPs granted with OD designation in Europe. patient number, follow-up duration and study centers.
This study aimed to enhance the knowledge on which ther­ The primary outcomes were classified into four cate­
apeutic areas having high unmet clinical needs attracted the gories: 1) patient-relevant outcomes were endpoints well
greatest number of ATMP developers to participate in. defined which could be measured objectively, such as mortal­
Furthermore, the current development status of the ATMPs ity and overall survival for oncology, 2) patient-reported out­
was investigated to gain a preliminary perspective on the comes (PROs) were any reports of the status of a patient’s
products representing high promises for market launch in health condition originating directly from the patient, such as
the near future. Finally, pivotal clinical trial methodology for the Health Related Quality of Life (HRQoL), 3) clinician-
ATMPs that are currently in the late-stage of development reported outcomes (ClinROs) were considered as rating scales
were studied to understand potential challenges faced in of patients’ status conducted on the basis of clinicians’ evalua­
clinical evidence collection for ATMPs. tion, and 4) surrogate outcomes were endpoints used to pre­
dict clinical benefits and support faster market approval, such
as biomarkers.
2. Method Two analysts conducted independent searches and extrac­
2.1. Data source tions of ODD-ATMP information. Any discrepancies identified
were resolved by a senior researcher specialized in pharma­
The official website of the European Commission (EC) was ceutical science and biologics.
searched to identify designated ODs as of 1 March 2020. The
official website of the European Medicines Agency (EMA) was
2.2.2. Sponsor-related data
searched to obtain the public summary reports on orphan
Sponsor-related data extracted included: sponsor name, spon­
designation published by the Committee for Orphan
sor type (commercial or noncommercial organization), and
Medicinal Products (COMP), as well as the scientific recom­
sponsor regions. Commercial organizations referred to phar­
mendations of ATMPs published by the Committee for
maceutical companies (including biotech company), consult­
Advanced therapies (CAT) [17].
ing companies, contract research companies or other science
The company websites and press releases for ATMPs spon­
companies. Noncommercial organizations referred to the fol­
sors were searched to understand sponsor characteristics and
lowing categories: private individual, medical centers (includ­
the current development status of their products. The United
ing hospitals), academic institutions (e.g., universities), charity
States National Library of Medicine’s ClinicalTrials.gov data­
organizations, and governmental agencies. The EMA-small and
base and the European Union’s (EU) Clinical Trials Register
medium-sized enterprises (SMEs) database was searched to
were searched to supplement information where information
identify whether a company held an SME status in the EU.
related to clinical trials of ATMPs was not completely pre­
For pharmaceutical companies originating outside of the
sented on the company websites.
EU, a legal representative in one of the EU Member States
(including the United Kingdom (UK)) must be established for
2.2. Data extraction and analysis administrative reasons. Therefore, both regions for ODs
holders and regions for original ODs developers were studied.
Specific extraction forms were established using Microsoft Parent organization regions were considered if the ODs
Excel to extract data. holders were subsidiary companies.

2.2.1. Product-related data


Product-related data including product name, product cate­ 3. Results
gory, cell type, gene vector, indication, prevalence of target
3.1. Description of orphan ATMPs and rare diseases
rare disease in EU, available treatment authorized, date of ODs
targeted
designations, and current development phase were searched
and extracted. The ATMPs were classified based on regulation A total of 1,670 products were designated as ODs as of
EC No. 1394/2007, under which, gene-modified, cell-based March 2020. Among them, 274 products met the definition
therapy, including chimeric antigen receptor T (CAR-T) immu­ of ATMPs and were investigated in this study. The number of
notherapy, was defined as GTMPs. ATMPs has significantly increased over the past 10 years: there
The therapeutic areas of targeted diseases were identified by were only 28 ATMPs between 2001 and 2009 in comparison to
searching the International Classification of Disease (ICD)-11 data­ 243 ATMPs between 2010 and 2019 (Supplementary
base. Diseases were classified as debilitating diseases, life- Material 1).
threatening diseases, or debilitating and life-threatening diseases GTMPs (N = 160, 58.39%) represented the largest number
according to the description of rare diseases in the COMP reports. of ATMPs, most of them used adeno-associated virus (AAV)
EXPERT OPINION ON ORPHAN DRUGS 419

(N = 110, 68.75%), lentiviral (N = 29, 18.13%) or retroviral origin of ODDs developers, 156 (56.93%) ATMPs were origin­
(N = 11, 6.88%) as gene vector. Among them, four products ally developed by European Member States. Among these,
adopted gene editing technology, including EDIT-101 (CRISPR/ France (28, 17.95%) held the largest number of ATMPs, fol­
Cas9-based), SB-318 (zinc finger nucleases-based), SB-913 (zinc lowed by England (27, 17.31%) and Germany (21, 13.46%)
finger nucleases-based), and CTX-001 (CRISPR/Cas9-based). (Figure 1). A total of 115 (41.97%) ATMPs were developed by
Five products, BB2121, JCAR017, KTE-X19, CTL019, and sponsors originating outside the EU, with over three-quarters
SLAMF7, were CAR-T cell-based gene therapy. sCTMPs of manufacturers from the United States (N = 91, 79.13%).
(N = 107, 39.05%) accounted for a relatively smaller percen­ Other countries with manufacturers having ATMPs in Europe
tage compared with GTMPs, with the majority of them (55, included Switzerland (N = 11), Japan (N = 4), Israel (N = 3),
51.40%) manufactured using allogeneic cell. TEPs represented India (N = 2), South Korea (N = 2), Canada (N = 1), and
the smallest number of ODDs-ATMPs (N = 7, 2.55%). Australia (N = 1).

3.1.1. Country distribution of ATMPs developers 3.1.2. Characteristics of ATMPs holders


Considering ODs holders in the EU, England (N = 69, 25.18%) The majority (N = 243) of ATMPs holders were commercial
had the largest number of ATMPs, followed by France (N = 43, organizations, only a small number (N = 31) of ATMPs holders
15.69%) and Germany (N = 29, 10.58%). When considering the were noncommercial organizations Figure 2. For 156 ATMPs

Figure 1. Number of ATMPs in member states of European Union.

Figure 2. Classification of ATMPs holders (data in percentage).


420 T. QIU ET AL.

originally developed in EU Member States, 125 (80.13%) of a narrow patient group. Twenty-three (8.39%) ATMPs were
them were developed by commercial organizations, and 74 indicated for diseases only having symptom-relieving treat­
(59.20%) of these companies were registered as SMEs in ments available. Thirty-one (11.31%) ATMPs were indicated
the EU. for diseases with replacement treatments available, such as
The companies or agencies with more than 5 orphan plasma factor for hemophilia, and alglucosidase alfa for glyco­
ATMPs included Promethera Biosciences (15 designations for gen storage disease type II.
9 indications), Fondazione Telethon (9 designations for 8 indi­ The therapeutic areas with the largest number of ATMPs
cations), Kite Pharma (7 designations for 7 indications), were metabolic diseases (64 designations for 33 diseases),
Orchard Therapeutics (7 designations for 6 indications), ophthalmology (41 designations for 12 diseases), oncology
UniQure Biopharma (6 designations for 5 indications), (28 designations for 7 diseases), neurology (25 designations
Abeona Therapeutics (5 designations for 4 indications), for 11 diseases), and hematologic malignancies (25 designa­
MeiraGTx (5 designations for 4 indications), and Novartis tions for 12 diseases) (Supplementary Material 2).
Europharm (5 designations for 5 indications). The diseases holding the greatest number of ATMPs were
retinitis pigmentosa, followed by epidermolysis bullosa,
glioma, Duchenne muscular dystrophy, and graft-versus-host
3.1.3. Target therapeutic areas of ATMPs disease Table 2.
A total of 116 types of rare diseases were targeted by 274
ATMPs. Among them, 158 (57.66%) ATMPs were indicated for
diseases perceived as debilitating and life-threatening diseases
Table 1. The greatest number (N = 126, 45.99%) of ATMPs 3.2. Current development stages of ATMPs
were indicated for diseases with a prevalence of 0.1 ~ 1/10,000 A total of 12 ATMPs for 16 rare diseases have received MA
people in the EU Table 1. worldwide of all the ATMPs investigated. Seven products were
Seventy-three (26.64%) ATMPs were indicated for diseases approved in the EU, including darvadstrocel (Alofisel®), auto­
currently having no authorized treatment available, while logous CD34+ cells encoding βA-T87Q-globin gene
patients were only treated with surgery, or other supportive (Zynteglo®), tisagenlecleucel (Kymriah®), axicabtagene ciloleu­
strategies. Thirty-three (12.04%) ATMPs were indicated for cel (Yescarta®), voretigene neparvovec (Luxturna®), ex vivo
diseases having authorized, but not satisfactory treatments expanded autologous human corneal epithelial cells contain­
available, such as in the case where treatments only targeted ing stem cells (Holoclar®), and autologous CD34+ cells trans­
duced with a lentiviral vector containing the human ADA gene
(Strimvelis®). Five products were approved in other countries
Table 1. Characteristic of targeted rare diseases of ATMPs.
outside of Europe, including remestemcel-L (Prochymal®) in
Disease
Therapeutic areas Disease characteristic* prevalence Existing Canada, autologous mesenchymal stem cell (MSC)
(N) (N) (N) treatments (N) (NeuroNata-R®) in South Korea, autologous adult live-
Metabolic Life-threatening disease ≤ 0.1 (64) No authorized cultured osteoblasts (Ossgrow®) as well as allogeneic bone
diseases (64) (19) treatment marrow derived MSC (Stempeucel®) in India, and Zolgensma®
(73)
Ophthalmological Debilitating disease (58) > 0.1–1 Not satisfactory in the US. Allogeneic adipose-derived adult MSC contained in
diseases (41) (126) treatment a fibrin-based bioengineered dermis (SkinMed) was used
(33) under the ‘Hospital Exemption’ rule in the EU. One anti-
Oncological Debilitating and life- > 1–2.5 Symptomatic-
diseases (28) threatening disease (26) relieving Epstein-Barr virus (EBV) cytotoxic T-cell therapy, developed
(158) treatment by Scotland National Health Services, was accepted by the
(23) Medicine and Healthcare Products Regulatory Agency
Neurological Debilitating and > 2.5 (58) Replacement
diseases (25) potentially life- therapy (31) (MHRA) for the application of this unlicensed medicinal pro­
threatening disease duct under the direct personal responsibility of the prescriber.
(30) The development of four products, INGN 201, ATIR101,
Hematologic Disability and life Authorized
Malignancies threatening disease (8) therapy SAR421869, and SAR422459, were terminated due to re-
(25) (satisfactory) prioritization of clinical development strategies. Alipogene
(99) tiparvovec (Glybera®) was withdrawn from the EU market
Immunological Negative impact on the Neutralizer or
diseases (21) QoL (1) chelating when the manufacturer did not apply for MA renewal.
agent (15) Human heterologous liver cells (Heparesc®) and sitimagene
Blood diseases ceradenovec (Cerepro®) were rejected for MA from EMA due
(21)
Dermal diseases to insufficient efficacy evidence.
(13) The majority of ATMPs (N = 192, 70.07%) were still in the
Transplantation- early development stages: 60 (21.90%) in pre-clinical phase, 12
related (8)
Digestive diseases (4.38%) in phase I clinical trials, 87 (31.75%) in phase I/II clinical
(6) trials, and 33 (12.04%) in phase II clinical trials. Only 28
Infectious diseases (10.22%) ATMPs were in phase II/III or phase III clinical trials.
(5)
Other diseases Among them, all clinical trials were multi-center studies
(17) (Figure 3), with the exception of 2 clinical trials, EB-101 for
N number, QoL quality of life epidermolysis bullosa and FLT180a for hemophilia B, which
EXPERT OPINION ON ORPHAN DRUGS 421

Table 2. Rare diseases with more than 5 ATMPs granted by the European medicines agency.
Therapeutic ATMPs Disease Availability of
Disease name areas Number prevalencea treatment The ATMPs after pre-clinical development (Number of products)
Retinitis pigmentosa Ophthalmology 14 3 Luxturna® GS030; HORA-PDE68; HORA-RPE65; AAV-RPGR; NSR-RPGR; AAV2-hRPE65v2;
UshStat; RST-001; Expanded hRPC from neural retina (N = 9)
Epidermolysis bullosa Dermatology 11 0.5 None EB-101; KB103; SkinMed; Holostem: COL17A1, COL7A1 and LAMB3-based
therapy; Allogeneic dermal fibroblasts; Allogeneic skin-derived ABCB5-
positive mesenchymal stem cells (N = 8)
Glioma Oncology 11 2.6 Chemotherapies AV0113; DNX-240; ICT-107; VB-111; DCVax-L; Toca 511; Autologous ex-vivo-
expanded leucocytes treated with 5-aza-2ʹ-deoxycytidine (N = 7)
Duchenne muscular Neurology 8 0.5 Translarna® PF-06939926; SGT-001 (N = 2)
dystrophy
Graft-versus-host Immunology 8 0.5 Immuno- Allogeneic bone-marrow derived ex-vivo expanded multipotent adult
disease suppressants progenitor cells (N = 1)
Pancreatic cancer Oncology 7 2.4 Chemotherapies GVAX; LOAd703; VCN-01; Cytochrome P450 isoform 2B1 gene transfected
human embryonic kidney 293 cells encapsulated in polymeric cellulose
sulfate (N = 4)
Leber congenital Ophthalmology 6 1 Luxturna® AAV-RPE65; AAV2-hRPE65v2; EDIT-101; GS010 (N = 4)
amaurosis
Hemophilia B Hematology 6 0.3 Factor IX FLT180a; PF-06838435 (SPK-9001); AMT-061 (N = 3)
Hemophilia A Hematology 5 0.8 Factor VIII TAK-754; BMN 270; SPK-8011; SB-525 (N = 4)
Mucopolysaccharidosis Metabolic 5 0.1 None ABO-102; EGT-101; LYS-SAF302; SAF-301 (N = 4)
type IIIA disease
Ornithine Metabolic 5 0.1 Ravicti® DTX301; MRT5201 (N = 2)
transcarbamylase disease
b
Achromatopsia Ophthalmology 5 0.15 ~ 0.3 None AAV-CNGA3; ACHM A3; AAV-CNGB3; ACHM B3 (N = 4)
Severe combined Immunology 5 0.01 ~ 0.1b None OTL-101; Autologous CD34+ cells transfected with retroviral vector
immunodeficiency containing adenosine deaminase gene (N = 2)
ATMPs advanced therapy medicinal products, hRPC human retinal progenitor cell
Luxturna® Voretigene neparvovec, Ravicti® Glycerol phenylbutyrate, Translarna® Ataluren
aper 10,000 European citizens
bprevalence varied according to the disease subtypes.

were conducted in a single center in California and the UK, products were not as promising as anticipated: VB-111 for
respectively. recurrent glioblastoma did not demonstrate clinical benefits
Phase III studies were completed for four products, among in either overall survival or progression-free survival versus
them, GS010 showed clinically meaningful improvement in Bevacizumab group, Cx401 for complex perianal fistulas failed
vision against placebo in the 96 week cutoff data. However, to show significant difference in healing rate compared to the
the clinical results of the phase III study for the other three control group, and ECCS-50 for the treatment of impaired

Figure 3. Current status and study centers for phase II/III and phase III clinical trials.
422 T. QIU ET AL.

hand function in patients with systemic sclerosis missed both functional rating scale (ALSFRS-R) for NurOwn® and the
the primary and secondary outcomes s against placebo. Cochin score for ECCS-50. ClinROs were used for three pro­
Phase III studies were terminated for three products, auto­ ducts: Modified International Cooperative Ataxia Rating Scale
logous osteoblastic cells (Preob®) for non-traumatic osteone­ (mICARS) for Ery-Dex, Development quotient (DQ) for LYS-
crosis, ELAD for acute liver failure, and Toca 511 for glioma, all SAF302 and International Standards for Neurological
due to interim results suggesting that primary outcomes could Classification of Spinal Cord Injury (ISNCSCI) motor scores for
be unlikely to achieve in the final analysis. Phase III studies Neuro-Cells. Surrogate outcomes were used for 14 products: 1)
were suspended for two products, ICT-107 and BPX-501, progression-free survival or event-free survival for Lenti-D,
because the sponsors were unable to secure sufficient finan­ JCAR017, DCVax-L and BB2121, 2) treatment response rate
cial resources. for Preob®, Cx401, EB-101, and ATA129, 3) plasma factor activ­
The recruitment of phase III studies was not yet initiated for ity for BMN 270, FLT180a, SPK-9001 and AMT-061, and 4)
2 products, SPK-8011 and Neuro-Cells. SPK-8011 was being visual acuity for NSR-REP1 and GS010. The validity of primary
investigated in a lead-in, observational study prior to phase outcomes for two products, the time to neutrophils engraft­
III clinical trial. Neuro-Cells planned to start patient enrollment ment for NiCord® and the rate of change in Ellipsoid Zone (EZ)
in June 2020. Area Loss for NT-501, were not well established.
The recruitment of phase II/III or phase III studies for the
remaining 17 products is underway, with the exception of
autologous MSC-NTF cells (NurOwn®) having completed their 4. Discussion
recruitment with 261 patients included. The interim analyses
4.1. ATMPs potential to treat rare diseases lacking
of ongoing phase II/III or phase III studies were disclosed for
satisfactory therapy
five products, all of them showed encouraging results: Lenti-D
targeting adrenoleukodystrophy, JCAR017 targeting follicular The fast growth in the development of orphan ATMPs could
lymphoma, autologous dendritic cells (DCVax-L®) targeting be attributed to the benefits associated with ODs, such as the
glioma, AMT-061 targeting hemophilia B, and BMN 270 target­ guarantee of development support from the EMA and faster
ing hemophilia B. No interim data were released for the rest of patient access with high prices [18,19]. The UK and France
11 products. The MA application of BMN270 was submitted to were shown as the pioneering countries amongst the EU
EMA and FDA in the last quarter of 2019. Member States in this market [20]. The UK has been perceived
as the leading country with over 70 ATMPs companies actively
participating in this field. Biotech companies in France were
3.3. Study design of the pivotal studies for ATMPs
consistently strong contributors in ATMP development, such
The median patient size is 127 (ranging from 15 to 414), with as GenSight Biologics, Vivet Therapeutics, Lysogene and
12 (42.86%) studies having estimated enrollments less than TxCell [21].
100 patients. Among them, 15 (55.56%) studies were open- As shown in this study, around 50% of the ATMPs investi­
label studies, while the remaining 12 studies were blinded gated were indicated for rare diseases currently lacking effec­
(Table 3). tive treatments or having therapies unsatisfactory to patients’
Single-arm studies were conducted for 8 (29.63%) products needs. This suggested that orphan ATMPs in development
(Supplementary Material 3). Among them, BMN207, FLT180a, might provide new treatment opportunities for patients suf­
SPK-9001, and AMT-061 were indicated for hemophilia with fering from life-threatening rare diseases.
plasma factor replacement therapy available. Lenti-D, EB-101, A large number of ATMPs were used for the treatment of
LYS-SAF302, and ATA129 were indicated for diseases currently inherited metabolic disorders that mostly could be alleviated
having no available treatments. The natural history study of only by adherence to strict diet management. Enzyme repla­
mucopolysaccharidosis type IIIA was being conducted in parallel cement therapy (ERT) was applied for enzyme deficiency dis­
with the ongoing phase III studies investigating LYS-SAF302. eases, such as mucopolysaccharidos (MPS), toward supplying
Randomized clinical trials (RCTs) using active comparators the functional enzymes to accelerate the degradation of accu­
or standard-of-care (SoC) as control groups were conducted mulated toxic substances. Whereas ERT was associated with
for seven products: ELAD, BPX-501, omidubicel (NiCord®), persistent complications as well as the risk of developing anti-
Cx401, JCAR017, Toca511, and BB2121. Randomized, sham- drug antibodies to ERT, which could significantly affect clinical
controlled studies using placebo or untreated patients as outcomes [22]. The advantages of gene therapies for meta­
control groups were conducted for 11 products: Preob®, bolic diseases included their ability to deliver normal genes to
NurOwn®, Ery-Dex, NSR-REP1, ICT-107, VB-111 (for the treat­ generate therapeutic enzyme in sufficient quantities through
ment of glioma and ovarian cancer), DCVax-L, GS010, Neuro- one-time administration and their precise localization and
Cells, ECCS-50, and NT-501. The natural history study of rapid delivery to affected organs, including the central ner­
Leber’s hereditary optic neuropathy was being conducted in vous system (CNS) [23].
parallel with the ongoing phase III studies investigating Similar to enzyme deficiency diseases, hemophilia, as an
GS010. X-linked monogenic blood disorder, relies on frequent (every
Patient-relevant outcomes were used for five products: 2 ~ 3 days) coagulation factors replacement therapy to reduce
overall survival for ELAD, BPX-501, ICT-107, VB-111 (for the the incidence of spontaneous bleeding [24]. However, the
treatment of glioma and ovarian cancer), and Toca511. PROs formation of anti-drug antibodies has restricted the long-
were used for two products: Amyotrophic lateral sclerosis term effectiveness of such therapies. Encouragingly,
EXPERT OPINION ON ORPHAN DRUGS 423

Table 3. ATMPs (N = 28) being investigated in the phase II/III or phase III clinical trials.
Estimated
Product Patient completion Follow-up
name Indications Study design Comparator Primary outcomes numbera date duration
Preob® Non-traumatic Triple Placebo Percentage of treatment responders 55 Terminated 24 months
osteonecrosis blinded,
RCT
ELAD Acute liver failure Open label, Standard of care Overall survival 151 Terminated 91 days
RCT
BPX-501 HSCT Open label, CTX Overall survival 178 December 2023 3 years
RCT
NiCord® HSCT Open label, Unmanipulated Time to neutrophil engraftment 124 December 2019 42 days
RCT cord blood unit
Lenti-D Adrenoleukodystrophy Single arm NA Proportion of subjects who are alive and have 32 February 2023 24 months
trial no 6 major functional disabilities
NurOwn® ALS Double Placebo Amyotrophic lateral sclerosis functional rating 261 October 2020 28 weeks
blinded,
RCT
Cx401 Anal fistula Double Fibrin adhesive Closure of fistulas 214 Completed 26 weeks
blinded,
RCT
Ery-Dex Ataxia telangiectasia Triple Placebo Modified International Cooperative Ataxia 180 December 2019 12 months
blinded, Rating
RCT
NSR- Choroideremia Single Untreated Corrected visual acuity 169 March 2020 12 months
REP1 blinded, patients
RCT
EB-101 Epidermolysis bullosa Single arm NA Wound healing 15 March 2021 12 weeks
trial
JCAR017 Follicular lymphoma Open label, Standard of care Event free survival 262 May 2023 3 years
RCT
ICT-107 Glioma Double Placebo Overall survival 414 Suspended 46 months
blinded,
RCT
VB-111 Glioma Open label, Add on therapy Overall survival 252 June 2023 10 years
RCT
DCVax-L Glioma Quadruple Placebo Progression free survival 331 Unknown Unknown
blinded,
RCT
Toca 511 Glioma Open label, Standard of care Overall survival 403 December 2019 4 years
RCT
BMN 270 Hemophilia A Single arm NA Median FVIII activity 130 December 2022 52 weeks
trial
SPK- Hemophilia A NA NA NA 55 July 2020 12 months
8011
FLT180a Hemophilia B Single arm NA Factor IX activity levels 50 December 2035 5 years
trial
SPK- Hemophilia B Single arm NA Annualized bleeding rate; Vector derived FIX: 55 November 2021 12 months
9001 trial C level
AMT-061 Hemophilia B Single arm NA Factor IX activity levels 56 March 2020 26 weeks
trial
GS010 Leber’s hereditary optic Quadruple Placebo ETDRS visual acuity 37 Completed 48 weeks
neuropathy blinded,
RCT
LYS- SAF302 MPS type IIIA Single arm trial NA Development 20
quotient
January 2022 24 months
BB2121 Multiple myeloma Open label, Standard of care Progression free survival 381 June, 2025 5 years
RCT
VB-111 Ovarian cancer Double Placebo Overall survival 400 December 2022 5 years
blinded,
RCT
ATA129 Post-transplant Single arm NA Objective response rate 33 November 2020 2 years
lymphoproliferative
disorder
Neuro- Spinal cord injury Double Placebo Physical changes; Increase of ISNCSCI motor 70 January 2021 12 months
Cells blinded, scores
RCT
ECCS-50 Systemic sclerosis Quadruple Placebo Cochin score (24 weeks) 88 Completed 48 weeks
blinded,
RCT
NT-501 Macular telangiectasia Double Placebo Rate of Change in Ellipsoid Zone Area Loss 112 March 2022 24 months
type 2 blinded,
RCT
ALS amyotrophic lateral sclerosis, AML acute myeloid leukemia, ATMPs advanced therapy medicinal products, CTX cyclophosphamide, ETDRS Early Treatment Diabetic
Retinopathy Study, HSCT hematopoietic stem cell transplantation, ISNCSCI International Standards for Neurological Classification of Spinal Cord Injury, MPS
mucopolysaccharidosis, NA not applicable, RCT randomized controlled trial
NiCord® omidubicel, NurOwn® autologous-cultured mesenchymal bone marrow stromal cells secreting neurotrophic factors, Preob® autologous osteoblastic cells
aActual patient number was presented if the study has completed, otherwise, estimated patient number was presented.
424 T. QIU ET AL.

hemophilia was considered well suited for correction by gene This analysis has shown that surrogate outcomes were
therapy as the bleeding phenotype was seen to be responsive commonly selected as primary outcomes for ATMPs.
to a wide range of factor levels, thus a precise regulation was However, surrogate outcomes raised questions regarding
not required [25]. Valoctocogene roxaparvovec (BMN270) for their appropriateness to support MA [37]. It was advisable
hemophilia A has submitted a market authorization applica­ that the validity of surrogate outcomes be established early
tion to the EMA and FDA in November and December of 2019, before deciding to use surrogate outcomes in the pivotal
respectively, based on data from a phase 1/2 trial showing studies. PROs can provide powerful evidence for patient per­
reduced bleeding episodes and need for factor VIII infusions. ceptions that clinical measures are unable to capture [32].
Remarkably, the levels of clotting factor VIII remained stable Composite outcomes including multiple types of outcomes
over the course of 3 years following treatment [26]. could be considered to evaluate treatment outcomes for
Inherited retina diseases, such as retina pigmentosa and patients with heterogeneous disease characteristics [36,38].
Leber’s congenital amaurosis, also attracted a attention from Early engagement of patient groups could be applied to
ATMP developers. The retina has been considered an excellent capture the additional values of ATMPs [39].
target for gene therapy since the accessibility to the retina
allows for relatively noninvasive procedures, the treatment
4.2.2. Market access for ODs and ATMPs in Europe
effects can be easily monitored, and the blood–retinal barrier
The high prices for ODs, combined with the great uncertainties
could minimize the systemic immune response [27].
in clinical evidence, can result in the incremental cost-
Luxturna®, a treatment delivering a normal copy of the
effectiveness ratio (ICER) of ODs exceeding the payer ‘willing­
RPE65 gene directly to retinal cells, was approved as the first
ness to pay’ threshold. Several EU countries have established
therapy to treat patients with retinal dystrophies caused by
special pathways to overcome the challenges in the HTA of
‘biallelic’ RPE65 mutations, with the potential to restore sight
ODs. For example, ultra-ODs may be eligible to undergo Highly
and improve vison at low light levels as compared to the
Specialized Technology evaluations in England, which raises the
control group.
ICER threshold to £100,000 per QALY gained [40]. Approved
ODs were presumed to have additional benefit by law, granting
with reimbursement along with MA automatically in Germany
4.2. Challenges in the patient access to orphan ATMPs [41]. A variety of managed entry schemes is increasingly imple­
mented across the European countries to address the uncer­
4.2.1. Challenges in conducting clinical trials for ATMPs
tainties of ATMPs, ranging from the simple discount to
Despite extensive interests for developing orphan ATMPs, the
coverage with evidence development programs containing
number of ATMPs approved as well as the ATMPs in late stage
pay-for-performance elements [42].
of development globally is still limited. Small patient size
Although ATMPs may benefit from the above-mentioned
constitutes the biggest obstacles in conducting clinical trials
specific HTA pathways for ODs established in EU countries,
of ODs. Insufficient knowledge about rare diseases leads to
there remains inherent uncertainties unique to ATMPs. The dur­
a delay in correct diagnosis [28], thus candidates also miss the
ability of treatment benefits and potential risks in the long run is
ideal opportune time to be enrolled and treated earlier [29].
impossible to predict based on the limited evidence at the time
Placebo-controlled studies face ethical hurdles in rare diseases,
of market launch [36]. Traditional HTA pathways may fail to
while rare diseases usually lack commonly agreed upon SOC,
capture the broader societal values associated with ATMPs,
posing challenges to conduct RCTs with accepted active com­
including the age of onset (priority given to children), lifetime
parators. Heterogeneity in pathophysiology of rare diseases
disease burden for patients [13], productivity, improvement in
leads to great variations in drug responses [6]. This situation
adherence and the impact on the future drugs development
has become even more complex when it comes to ATMPs for
[42]. The recent sanitary COVID-19 crisis supports the additional
rare diseases, which generally require complicated manufac­
value of one-time therapies over chronic treatments on regular
turing processes. Maintaining the high quality and minimizing
basis in terms of reducing the contamination risk at hospital
the batch-to-batch variation of ATMPs are crucial to ensure
level. Further explorations are needed regarding how best to
adequate patient response [30]. Additionally, as one-time
measure and include the additional value elements into the
administration therapies, ATMPs will exclude the patients
QALY calculation would be informative and useful [43,44].
from eligibility in future clinical trials [31,32].
Innovative methods to strengthen the evidence quality for
ATMPs have been proposed [33]. Natural history study, exter­
4.3. Business Model for ATMPs in rare conditions
nal prospective cohort, or patient registry to serve as the
control group of the single-arm trial have been utilized to This study has indicated that the development of ATMPs was
gain regulatory approval. Pragmatic RCTs incorporating dominated by commercial sponsors rather than noncommer­
a cluster randomization design based on study centers [34] cial sponsors. This was partly because noncommercial organi­
may provide a robust design. Other strategies to deal with zations signed licensing agreements with pharmaceutical
small sample sizes include adaptive study design [35], employ­ industries to achieve the translation from scientific discoveries
ing genetic testing to reduce individual’s variability, and to ultimate commercialization. Public–private partnership
selecting continuous endpoints [29]. Moreover, data-sharing complementarily harnessed the capabilities of each party,
among multiple companies developing ATMPs for the same thus ensuring a faster development of ATMPs [45,46].
patient group could be a practical and ethical solution [36]. Biotech stocks have far outpaced the average stock market
EXPERT OPINION ON ORPHAN DRUGS 425

during the last quarter of 2019, following a 195 USD billion 5.2. Regulatory requirements
biopharma acquisition witnessed among pharmaceutical
The FDA predicts that they will be approving between 10 and
giants [47]. The rapid progress on ATMPs and the extensive
20 cell and gene therapy products a year based on an assess­
pipelines in biotech companies have urged interested big
ment of the current pipeline and the clinical success rates by
pharma to act quickly in order to catch the opportunity to
2025. Fast growth in ATMPs will enhance the knowledge
arrive early on the market.
about effectiveness and safety of ATMPs, thus regulators
Due to the paucity of clinical evidence, it is still prema­
may take advantage of insights gained from already approved
ture to predict the magnitude and sustainability of clinical
products to impose more clarified and strict requirements on
benefits of ATMPs in the long run. On the most optimistic
the collection of clinical evidence. The sustainability of the
assumptions, there are gene replacement therapies target­
effectiveness of ATMPs will be better understood, leading to
ing monogenic conditions may bring a ‘cure’ opportunity, or
requirements for clinical trial durations to be sufficiently long
at least, patients may have symptoms relieved for many
enough to investigate the patient outcomes. The require­
years after one administration. Under such circumstance,
ments for comparative studies may be more demanded in
once the limited number of prevalent patients have been
the existence of alternative therapies with reasonable efficacy,
presumably cured, only incident patients remain available to
such as chemotherapies for large B-cell lymphoma. Historical
treat. Therefore, future competitors must differentiate and
comparisons will be accepted only when rigorous methodolo­
highlight their respective clinical advantages as comparisons
gical protocols are in place to guarantee the validity of such
to initially approved products. Otherwise, they will probably
comparisons. Post-approval commitment for effectiveness evi­
end up with ‘no added benefits’ against existing product,
dence generation will be unavoidable. The difficulties inherent
left with no options but lower prices as the only leverage to
to all ODs development will continue to apply, and an increas­
win the already depleted market. In another scenario closer
ing number of ultra-orphan drugs targeting monogenic dis­
to reality, ATMPs would not all be ‘miracle’ to fundamentally
eases will likely emerge and may struggle to generate robust
reverse disease progression, but show great variations in
pre-approval evidence.
clinical benefits for heterogeneous ATMPs and diverse dis­
While all biopharmaceutical industries have been
eases. For example, CAR-T cell therapy will not be cure for
affected by the COVID-19 pandemic, many gene or cell
blood cancers, as shown in the clinical trials that 47% of
therapy companies, especially biotech and start-up compa­
patients receiving Yescarta® had a complete response.
nies, have been disrupted particularly heavily due to their
Under such circumstances, first ATMPs, therefore, will not
complex manufacturing, supply, and funding mechanisms.
exhaust all the prevalent patients and still leave rooms for
Research and preclinical development activities were slo­
following products with different modes of actions. Even if
wed down due to less than normal operation capacity.
products with better efficacy or less safety concerns reach
Patient recruitment become ever challenging in small com­
the market later on, they may take up the leading position
munities with ultra-rare diseases, where international
of available products.
recruitments were suspended and engagements with
patients for follow-up assessment also suffered [32]. Some
companies have implied that their launch plans were mod­
5. Future prospect
ified or postponed, with delays in regulatory approval and
5.1. Advancement in technology patient access to ATMPs expected. Close connections with
regulators will be helpful to timely communicate the rele­
ATMPs hold new clinical promises in the treatment of devast­
vant modifications made in the manufacturing process and
ing diseases through its unique mechanism of action, yet
clinical trial protocols. Regulators, such as the FDA, have
substantial challenges remain in the manufacturing of auto­
also claimed that their work to advance treatments for rare
logous ATMPs. It requires an individual approach for each
diseases and ensure continuity of care for people with rare
patient, raising barriers in terms of complex logistics, lengthy
diseases remain top priorities during the COVID-19 out­
wait times, and variable product potency [48]. Therefore,
break, along with their endeavors to establish ‘Rare
developing allogeneic products would represent benefits to
Disease Clinical Trial Networks’ as a global clinical trial
eliminate the complex manufacturing logistics and inconveni­
platform for a wide range of rare diseases [52].
ence caused by in-patient administration [49]. For instance,
Companies with more efficiency and resilience will demon­
allogeneic induced pluripotent stem cell (iPSC)-derivatives
strate their advantages to absorb the damage and get back on
have been suggested as a unique platform for human disease
track more quickly, while fragile, small biotech will need to re-
modeling, target validation, drug screening, preclinical assess­
assess their portfolio and prioritization strategies to decide where
ment, and development of personalized medicine [50]. The
to invest further [53].
progress in gene-editing technologies, such as CRISPR-CAS9,
ZINC Finger, and Transcription activator-like effector nucleases
(TALENs) have enabled the possibility of developing engi­
5.3. Reexamine the reimbursement of ATMPs
neered constructs to create universal cellular therapies to be
used as ‘off-the-shelf’ [51]. Additionally, gene-editing technol­ Historically, most ATMPs have succeeded to gain reimbur­
ogy represents the potential to reduce the off-target activities sement in EU countries. While ATMPs themselves, relying
by target and kill cancer cells while also distinguishing and solely on its innovative nature, may not constitute
preserving healthy cells [50]. a sufficient reason to justify their cost-effectiveness
426 T. QIU ET AL.

considering the significant evidence gaps in terms of their not restricting to manufacturers, regulators, and payers, but
‘curative’ nature and long-term advantages at the time of also inclusive of patient advocacy groups and clinicians, will
market launch. Rather, rare diseases featured with substan­ promote the obtention of support and expertise that are
tial unmet needs that are targeted by ATMPs remain the essential to succeed. Patients have expressed their willingness
primary drivers for favorable reimbursement decisions. to be in partnership with manufacturers throughout the whole
However, it must be recognized that ODs have triggered development lifecycle instead of passive participants of clinical
continuous debates surrounding whether the societal pre­ trials. The advantages of patient contribution in terms of
ference toward rare disease really exists, and to what deepening knowledge on the natural history of rare diseases,
extent the healthcare system should give priority to ODs facilitating collection of patient-relevant outcomes, informing
[41]. One example is Glybera® for ultra-rare condition the understanding of patient perception on the value of
named lipoprotein lipase deficiency (LPLD), which was ATMPs, and facilitating the long-term patient tracking must
approved based on the scarce evidence from single-arm be highlighted [56].
trials recruiting less than 20 patients. However, it was The level of commercial returns generated by ATMPs is
withdrawn from the EU market because its 1 USD million expected to be highly dependent upon what proposition (i.e
price caused extremely limited usage in major EU countries supports/services) is provided in addition to the product itself
where reimbursement was lacking. This is indicative that [57]. As multiple ATMPs will gain approval over the following
the price of orphan ATMPs must be determined by finding years, hospital systems are at risk of becoming overloaded
the right balance between medical needs, clinical benefits, with managing multiple training programs and administration
appropriate commercial returns, and healthcare affordabil­ protocols. It is valuable in having manufacturers and third-
ity [54]. party suppliers come together and align on standardized pro­
Furthermore, although the cost burden of existing cesses to reduce complexity for hospitals [58]. Strategies to
orphan ATMPs is likely to be relatively small in the short facilitate clinical adoption will be needed to ensure ATMPs are
term, the cumulative effects of multiple ATMPs expanding as easy as possible to integrate into healthcare delivery/
indications from rare diseases to more prevalent diseases patient pathway. Additionally, clinicians may need support to
has raised financial concerns for the long term. Not sur­ secure health claims for patient administration are correctly
prisingly, payer acceptance for the evidence limitations for and efficiently collected to satisfy the data requirements of
ATMPs may not be sustained in the long run. Payers will public and private health insurers.
further emphasize the importance of comprehensive evi­ Under the COVID-19 crisis, vulnerable patients suffering
dence collection to confirm the sustainability of treatment from rare diseases will face the difficult choice between risking
benefits, relative effectiveness profile, and the impact on COVID-19 exposure and postponing clinician visits. Clearly,
patient-relevant outcomes. Innovative payment mechan­ urgent actions are required to transform healthcare delivery
isms (e.g. outcome-based payment and install payment) and to update current systems by making the most use of
will be increasingly adopted to mitigate the financial risks digital technologies, such as the use of teleconference to
in relation with evidence limitations of orphan ATMPs, thus deliver remote healthcare services [59]. Manufacturers of
better informing on their respective strengths and draw­ ATMPs will need to have the right IT infrastructures in place
backs. Real-world evidence (RWE) collection will be increas­ to facilitate the digitization of documentation and the disse­
ingly appreciated to provide important insights into the mination of training materials. If implemented appropriately,
post-launch performances to bridge evidence gaps at time digital health may be impactful to improve the healthcare
of launch, to support future reimbursement decisions, and access for patients desiderating orphan ATMPs in the
to strengthen the implementation of outcomes-based pay­ future [60].
ments. Widespread adoption of RWE may only materialize
when its inherent shortfalls, including potential confound­
ing, incomplete data, lack of universally accepted metho­ 6. Conclusion
dological standards, have been overcome [55]. As shown in this study, ATMPs will bring substantial promises
In particular, affordability and sustainability of the to a variety of rare diseases currently out of treatment options.
healthcare system become more evident and urgent issues It is predicted that a rising number of orphan ATMPs will arrive
in the current COVID-19 pandemic, when constrained in the foreseeable future, with some products currently in the
healthcare resources must be prioritized to combat this late stages of clinical trials. However, challenges in the gen­
catastrophic emergency. More than ever before, ATMP eration of compelling and robust evidence for orphan ATMPs
developers will need to demonstrate that the costs of cannot be neglected due to small patient numbers, open-label
their treatments are offset by long-term, measurable designs, absence of suitable comparators, inappropriate surro­
patient benefits [53]. gate outcomes, and short follow-up durations. Regulators tend
to embrace orphan ATMPs through initiatives of accelerated
approval pathways eligible for ODs and breakthrough thera­
5.4. Patient engagement and clinical adoption
pies. Conversely, payers scrutinize the high price of ATMPs
Collaborations involving all relevant stakeholders are of para­ making the cost-effectiveness unjustified in relation to the
mount importance for orphan ATMPs. Early communication, evidence uncertainties surrounding long-term performances.
EXPERT OPINION ON ORPHAN DRUGS 427

Undoubtfully, with ATMPs targeting both rare diseases and References


prevalent diseases emerging, more strict evidence require­
Papers of special note have been highlighted as either of interest (•) or of
ments will become a necessity for both regulators and payers. considerable interest (••) to readers.
The rising number of ATMPs competing in a niche market will 1. Gammie T, Lu CY, Babar ZU. Access to orphan drugs:
require developers to highlight the respective therapeutic a comprehensive review of legislations, regulations and policies
advantages of their products, especially for the products in 35 countries. PLoS One. 2015;10(10):e0140002.
launched later on. The success of orphan ATMPs will also •• This paper provided a comprehensive introduction about the
global regulations for orphan drugs regarding the legislations,
rely on collaborative strategies to streamline market launch incentives for manufacturers and reimbursement policies.
and commercialization processes. Public–private partnerships 2. Khosla N, Valdez R. A compilation of national plans, policies and
and cooperative agreements between biotechs and major government actions for rare diseases in 23 countries. Intractable
pharmaceutical companies could strengthen the capacities of Rare Dis Res. 2018;7(4):213–222.
each party and increase the efficiency of developing ATMPs. 3. Benjamin K, Vernon MK, Patrick DL, et al. Patient-reported outcome
and observer-reported outcome assessment in rare disease clinical
The early engagement of relevant stakeholders, including trials: an ISPOR COA emerging good practices task force report.
patients, clinicians, regulators, and payers, may be paramount Value Health. 2017;20(7):838–855.
in order to understand the needs of each one, and therefore, 4. Jayasundara K, Hollis A, Krahn M, et al. Estimating the clinical cost
enable developers to be better prepared to remove potential of drug development for orphan versus non-orphan drugs.
barriers. Additionally, collaborations and communications are Orphanet J Rare Dis. 2019;14(1):12.
5. Lacaze P, Millis N, Fookes M, et al. Rare disease registries: a call to
evermore critical during the COVID-19 outbreak when action. Intern Med J. 2017;47(9):1075–1079.
research and development activities for ATMPs have been 6. Sun W, Zheng W, Simeonov A. Drug discovery and development for
heavily disrupted. The benefits of a digital revolution to trans­ rare genetic disorders. Am J Med Genet A. 2017;173(9):2307–2322.
form the paradigm of healthcare deliver must be amplified to 7. Hanna E, Remuzat C, Auquier P, et al. Advanced therapy medicinal
ensure that timely patient access to orphan ATMPs will not be products: current and future perspectives. J Mark Access Health
Policy.. 2016;4(1):31036.
severely delayed. • This paper characterised the ATMPs in development and dis­
cuss future implications in terms of market access by searching
clinical trials registered in the EudraCT, ClinicalTrails.gov and
ICTRP.
Author contributions 8. Food and Drug Administration. Regenerative medicine advanced
therapy designation. 2019. cited 2020 Apr 28.Available from:
M Toumi, TT Qiu and E Hanna: contributed to the study conception and
https://www.fda.gov/vaccines-blood-biologics/cellular-gene-
design; TT Qiu, YT Wang, R Han and SY Liang: material preparation,
therapy-products/regenerative-medicine-advanced-therapy-
data collection and analysis. TT Qiu, M Toumi and M Dabbous: wrote
designation.
the first draft of the manuscript. All authors commented on previous
9. Qiu T, Dabbous M, Chachoua L, et al. The diversity in regenerative
versions of the manuscript. All authors read and approved the final
medicines regulations in Europe, USA and Japan. Cell Gene Ther
manuscript.
Insights. 2019;5(8):1031–1042.
10. Coherent Market Insights. Global cell and gene therapy market to
surpass US$ 35.4 Billion by 2026. Globe newswire. 2019. cited 2020
Mar 14. Available from: http://www.globenewswire.com/newsre
Declaration of interest lease/2019/02/05/1710761/0/en/Global-Cell-and-Gene-Therapy-
The authors have no relevant affiliations or financial involvement with any Market-to-Surpass-US-35-4-Billion-by-2026.html
organization or entity with a financial interest in or financial conflict with 11. Korchagina D, Millier A, Vataire A-L, et al. Determinants of orphan drugs
the subject matter or materials discussed in the manuscript. This includes prices in France: A regression analysis. Orphanet J Rare Dis. 2017;12
employment, consultancies, honoraria, stock ownership or options, expert (1):75
testimony, grants or patents received or pending, or royalties. 12. Jayasundara K, Hollis A, Krahn M, et al. Estimating the clinical cost
of drug development for orphan versus non-orphan drugs.
Orphanet J Rare Dis. 2019;14(1):12.
Funding 13. Grace Hampson AT, Pearson SD, Dreitlein WB, et al. Gene therapy:
evidence, value and affordability in the US health care system.
This paper was not funded. J Comp Eff Res. 7(1): 15–28. 2018.
•• Gene therapies present exciting opportunities, but also pose
major challenges in terms of evidence generation, value
assessment and affordability.
14. Ronfard V, Vertes AA, May MH, et al. Evaluating the past, present,
Reviewer Disclosures
and future of regenerative medicine: a global view. Tissue Eng Part
Peer reviewers on this manuscript have no relevant financial or other B Rev. 2017;23(2):199–210.
relationships to disclose. 15. Wurster CD, Ludolph AC. Nusinersen for spinal muscular atrophy.
Ther Adv Neurol Disord.. 2018;11: 1756285618754459.
16. Pipes SC How this gene therapy drug earned its $2.1 million price
ORCID tag. 2020. cited 2020 Apr 28. Available from: https://fortune.com/
2020/02/07/zolgensma-high-drug-prices/
Shuyao Liang http://orcid.org/0000-0002-0627-8922 17. European Medicines Agency. Committee for orphan medicinal pro­
Mondher Toumi http://orcid.org/0000-0001-7939-7204 ducts (COMP). 2020. cited 2020 Apr 28. Available from: https://www.
428 T. QIU ET AL.

ema.europa.eu/en/committees/committee-orphan-medicinal- 38. Nestler-Parr S, Korchagina D, Toumi M, et al. Challenges in research


products-comp and health technology assessment of rare disease technologies:
18. Medic G, Korchagina D, Young KE, et al. Do payers value rarity? report of the ISPOR rare disease special interest group. Value
An analysis of the relationship between disease rarity and Health. 21(5): 493–500. 2018.
orphan drug prices in Europe. J Mark Access Health Policy. •• Research-related challenges linked to the rare diseases were
2017;5(1):1299665. categorized into those pertaining to disease recognition and
19. Farkas AM, Mariz S, Stoyanova-Beninska V, et al. Advanced therapy diagnosis, evaluation of treatment effect, and patient recruit­
medicinal products for rare diseases: state of play of incentives ment for clinical research. HTA-related challenges were classi­
supporting development in Europe. Front Med (Lausanne). fied into issues relating to the lack of a tailored HTA method
2017;4:53. for orphan drugs and uncertainty for HTA agencies and health
20. Alliance for Regenerative Medicine. Leading innovation-the UK’s care payers.
ATMP landscape. 2019. 39. Mercieca-Bebber R, King MT, Calvert MJ, et al. The importance of
21. BIOTECH S. Why France is leading the gene and cell therapy field in patient-reported outcomes in clinical trials and strategies for future
Europe. 2019. cited 2019 Dec 14. Available from: https://www. optimization. Patient Relat Outcome Meas.. 2018;9:353–367.
sirion-biotech.com/newsroom_post/why-france-is-leading-the- 40. Czech M, Baran-Kooiker A, Atikeler K, et al. A review of rare disease
gene-and-cell-therapy-field-in-europe/. policies and orphan drug reimbursement systems in 12 Eurasian
22. Fraldi A, Serafini M, Sorrentino NC, et al. Gene therapy for muco­ countries. Front Public Health. 2019;7:416.
polysaccharidoses: in vivo and ex vivo approaches. Ital J Pediatr. 41. Young KE, Soussi I, Hemels M, et al. A comparative study of orphan
2018 Nov 16;44(Suppl 2):130. drug prices in Europe. J Mark Access Health Policy. 2017;5
23. Mark A Kay SLW, Woo SLC. Gene therapy for metabolic disorders. (1):1297886.
Trends Genet. 1994;10(7):253–257. 42. David Tordrup VT, Kanavos P. Orphan drug considerations in health
24. Batty P, Lillicrap D. Advances and challenges for hemophilia gene technology assessment in eight european countries. Rare dis
therapy. Hum Mol Genet. 2019;28(R1):R95–R101. orphan drugs. 2014;1(3):86–97.
25. Perrin GQ, Herzog RW, Markusic DM. Update on clinical gene 43. Lakdawalla DN, Doshi JA, Garrison LP, et al. Defining elements of
therapy for hemophilia. Blood. 2019;133(5):407–414. value in health care-a health economics approach: an ISPOR special
26. Today HN BioMarin seeks marketing approval in Europe for its task force report. Value Health. 2018;21(2):131–139.
investigational gene therapy, valoctocogene roxaparvovec. 2020. 44. Drummond MF, Neumann PJ, Sullivan SD, et al. Analytic considera­
cited 2020 Jul 29. Available from: https://hemophilianewstoday. tions in applying a general economic evaluation reference case to
com/2019/11/25/biomarin-submits-maa-to-ema-for-valoctocogene gene therapy. Value Health. 2019;22(6):661–668.
-roxaparvovec/ 45. Bubela T, Bonter K, Lachance S, et al. More haste, less speed: could
27. Ziccardi L, Cordeddu V, Gaddini L, et al. Gene therapy in retinal public-private partnerships advance cellular immunotherapies?
dystrophies. Int J Mol Sci. 2019;20(22):5722. Front Med (Lausanne). 2017;4:134.
28. Augustine EF, Adams HR, Mink JW. Clinical trials in rare disease: 46. Jaroslawski S, Toumi M. Non-profit drug research and develop­
challenges and opportunities. J Child Neurol. 2013;28 ment: the case study of Genethon. J Mark Access Health Policy.
(9):1142–1150. 2019;7(1):1545514.
29. Gagne JJ, Thompson L, O’Keefe K, et al. Innovative research meth­ 47. Investor’s Business Daily. Biotech stocks burn a hole in big phar­
ods for studying treatments for rare diseases: methodological ma’s pocket; look for more mergers in 2020. 2019. cited 2020 Apr
review. BMJ. 2014;349(nov24 15): g6802. 15. Available from: https://www.investors.com/news/technology/
•• Innovative clinical trial methods minimize sample size require­ biotech-stocks-catch-fire-big-pharma-opens-wallet/
ments and maximize the proportion of patients who receive 48. Chang D. Innovating CAR T cell therapy for today and tomorrow.
active treatment, strategies crucial to studying rare diseases Cell Gene Ther Insights. 2020;6(6):783–788.
with limited treatment choices. 49. Liu C, Wu J. The rise of human induced pluripotent stem cell
30. Abou-El-Enein M, Elsanhoury A, Reinke P. Overcoming challenges technology in drug development. Cell Gene Ther Insights. 2020;6
facing advanced therapies in the EU market. Cell Stem Cell. 2016;19 (1):127–135.
(3):293–297. 50. Bailey SR, Maus MV. Gene editing for immune cell therapies. Nat
31. Kilix S. Trends in cell and gene therapy clinical development for Biotechnol. 2019 Dec 01;37(12):1425–1434.
rare and ultra-rare diseases. Cell Gene Ther Insights. 2020;6 51. Ashmore-Harris C, Fruhwirth GO. The clinical potential of gene
(3):543–547. editing as a tool to engineer cell-based therapeutics. Clin Transl
32. De Castro Lopez MJ. Advanced therapy clinical trials for rare pedia­ Med. 2020 Feb 07;9(1):15.
tric disorders: key challenges and lessons learned. Cell Gene Ther 52. Food and Drug Administration. Rare disease therapy development
Insights. 2020;6(3):511–515. and access remain top FDA priorities during COVID-19 2020. 2020.
33. Potter BK, Khangura SD, Tingley K, et al. Translating rare-disease cited 2020 Aug 20. Available from: https://www.fda.gov/news-
therapies into improved care for patients and families: what are the events/fda-voices/rare-disease-therapy-development-and-access-
right outcomes, designs, and engagement approaches in remain-top-fda-priorities-during-covid-19
health-systems research? Genet Med. 2016;18(2):117–123. 53. McKinsey. COVID-19 and cell and gene therapy: how to keep
34. Abrahamyan L, Feldman BM, Tomlinson G, et al. Alternative designs innovation on track 2020. 2020. cited 2020 Aug 20. Available
for clinical trials in rare diseases. Am J Med Genet C Semin Med from: https://www.mckinsey.com/industries/pharmaceuticals-and-
Genet. 2016;172(4):313–331. medical-products/our-insights/covid-19-and-cell-and-gene-therapy
35. Food and Drug Administration. Adaptive designs for clinical trials -how-to-keep-innovation-on-track.
of drugs and biologics 2019. 2019. cited 2020 Feb 10. Available 54. Tarnowski J, Krishna D, Jespers L, et al. Delivering advanced thera­
from: https://www.fda.gov/regulatory-information/search-fda- pies: the big pharma approach. Gene Ther. 2017 Sep;24(9):593–598.
guidance-documents/adaptive-design-clinical-trials-drugs-and- 55. Hampson G, Towse A, Dreitlein WB, et al. Real-world evidence for
biologics-guidance-industry coverage decisions: opportunities and challenges. J Comp Eff Res..
36. Zheng PP, Kros JM, Approved LJ. CAR T cell therapies: ice bucket 2018 Dec;7(12):1133–1143.
challenges on glaring safety risks and long-term impacts. Drug 56. White W. A rare disease patient/caregiver perspective on fair pricing
Discov Today. 2018;23(6):1175–1182. and access to gene-based therapies. Gene Ther. 2019 Nov 13.Epub
37. Kim HT, Armand P. Clinical endpoints in allogeneic hematopoietic ahead of print. PMID:31723211.DOI:10.1038/s41434-019-0110-7.
stem cell transplantation studies: the cost of freedom. Biol Blood • From the patient perspective, current HTA value assessment
Marrow Transplant. 2013;19(6):860–866. frameworks disadvantage people with disabilities, may not
EXPERT OPINION ON ORPHAN DRUGS 429

accurately reflect patients’ and societal views, and omit neces­ •• This article focused on innovative immuno-oncology cell thera­
sary factors such as impact on caregivers, lost productivity, pies and explores key opportunities and challenges facing
and the future value of innovation. biotech companies, commercial plans to drive success, and
57. Malik NN, Durdy MB. CAR T-cell therapies: formulation-product- strategies to disrupt the market.
proposition framework for commercialisation. Drug Discov Today. 59. Keesara S, Jonas A, Schulman K. Covid-19 and health care’s
2016 Nov;21(11):1731–1734. digital revolution. N Engl J Med. 2020 Jun 04;382(23):e82.
58. Firestone G. Immuno-oncology cell therapies: commercial considera­ 60. Whitelaw S, Mamas MA, Topol E, et al. Applications of digital
tions and strategies for the new decade. Cell Gene Ther Insights. 2020;6 technology in COVID-19 pandemic planning and response.
(6): 798. Lancet Digital Health. 2020;2(8):e435–e440.

You might also like