Skip to main content
Tissue Engineering. Part B, Reviews logoLink to Tissue Engineering. Part B, Reviews
. 2023 Apr 10;29(2):167–187. doi: 10.1089/ten.teb.2022.0033

The Ethical Implications of Tissue Engineering for Regenerative Purposes: A Systematic Review

Anne-Floor J de Kanter 1,, Karin R Jongsma 1, Marianne C Verhaar 2, Annelien L Bredenoord 1,3
PMCID: PMC10122262  PMID: 36112697

Abstract

Tissue Engineering (TE) is a branch of Regenerative Medicine (RM) that combines stem cells and biomaterial scaffolds to create living tissue constructs to restore patients' organs after injury or disease. Over the last decade, emerging technologies such as 3D bioprinting, biofabrication, supramolecular materials, induced pluripotent stem cells, and organoids have entered the field. While this rapidly evolving field is expected to have great therapeutic potential, its development from bench to bedside presents several ethical and societal challenges. To make sure TE will reach its ultimate goal of improving patient welfare, these challenges should be mapped out and evaluated. Therefore, we performed a systematic review of the ethical implications of the development and application of TE for regenerative purposes, as mentioned in the academic literature. A search query in PubMed, Embase, Scopus, and PhilPapers yielded 2451 unique articles. After systematic screening, 237 relevant ethical and biomedical articles published between 2008 and 2021 were included in our review. We identified a broad range of ethical implications that could be categorized under 10 themes. Seven themes trace the development from bench to bedside: (1) animal experimentation, (2) handling human tissue, (3) informed consent, (4) therapeutic potential, (5) risk and safety, (6) clinical translation, and (7) societal impact. Three themes represent ethical safeguards relevant to all developmental phases: (8) scientific integrity, (9) regulation, and (10) patient and public involvement. This review reveals that since 2008 a significant body of literature has emerged on how to design clinical trials for TE in a responsible manner. However, several topics remain in need of more attention. These include the acceptability of alternative translational pathways outside clinical trials, soft impacts on society and questions of ownership over engineered tissues. Overall, this overview of the ethical and societal implications of the field will help promote responsible development of new interventions in TE and RM. It can also serve as a valuable resource and educational tool for scientists, engineers, and clinicians in the field by providing an overview of the ethical considerations relevant to their work.

Impact statement

To our knowledge, this is the first time that the ethical implications of Tissue Engineering (TE) have been reviewed systematically. By gathering existing scholarly work and identifying knowledge gaps, this review facilitates further research into the ethical and societal implications of TE and Regenerative Medicine (RM) and other emerging biomedical technologies. Moreover, it will serve as a valuable resource and educational tool for scientists, engineers, and clinicians in the field by providing an overview of the ethical considerations relevant to their work. As such, our review may promote successful and responsible development of new strategies in TE and RM.

Keywords: tissue engineering, regenerative medicine, bioethics, responsible innovation, social impact, scientific integrity

Introduction

Tissue Engineering (TE) uses biomaterials, (stem) cells and bioactive molecules to construct functional, living three-dimensional (3D) tissues in a laboratory environment (in vitro) or directly in a patient's body (in situ). Examples include tissue engineered skin, 3D bioprinted bone, and liver organoids for transplantation. TE is one of three pillars of the broader field of Regenerative Medicine (RM), which aims to “replace or regenerate human cells, tissue, or organs to restore or establish normal function.”1 Stem cell (SC) transplantation and gene transfer (GT) constitute the other two pillars.

As a rapidly evolving medical technology that has been argued to be potentially disruptive2 and even paradigm-changing,3 TE holds great promise for novel cures, but also requires attention to the ethical implications of its development, clinical translation, and future impact on individuals and society. Addressing such ethical implications early on and embedding societal values into the design of TE products will help TE to reach its ultimate goal of improving patient welfare.4

The TE field has been maturing over the last two decades and ethical issues have received attention of ethicists, social scientists, and ethics-minded biomedical scientists and engineers. This body of literature has last been reviewed comprehensively in 2008, at which time the field was still in its infancy.5 Since then, the first applications have reached the stage of clinical application.6,7 Emerging technologies such as 3D bioprinting, biofabrication, supramolecular materials, induced pluripotent stem cells (iPSCs), and organoids have entered the TE field, and a shift has taken place toward strategies that mobilize the inherent regenerative capacity of the human body–and its endogenous cell populations–to a greater extent than before (in situ TE).2,8

Therefore, we have performed a systematic review (SR) of the ethical implications of the development and application of TE for regenerative purposes, as mentioned in the academic literature. While TE might have applications outside RM (e.g., as a drug screening platform or for in vitro meat production), we have focused on the clinical application of TE for tissue regeneration. To our knowledge, this is the first time that the ethical implications of this field have been reviewed in a systematic manner. An SR has the advantage of being comprehensive, methodologically rigorous and transparent, and of reducing (selection) bias.9

Method

We performed an SR to identify the ethical implications of the development and application of TE for regenerative purposes. The reviewing process was performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for SRs10 and the adapted PRISMA ethics guidelines by Kahrass et al.9 Ethical implications were broadly understood to be expressions of an ethical or societal issue or consideration, which may but need not take the form of a fully argument-based reason.11

Search strategy

Four databases (Embase, PubMed, Scopus, and PhilPapers) were searched for relevant English language articles using a search string combining search terms for (a) TE, RM, or bioprinting with (b) ethics or human rights, covering the period January 01, 2008 until October 05, 2021. An experienced librarian was consulted regarding the design of the search strings and choice of databases to ensure that relevant keywords were included, and that a comprehensive area of biomedical, bioengineering, and ethics literature was covered. The original search was performed on March 30, 2020 and updated on October 05, 2021. See Supplementary Data S1 for the complete search strategy of all databases.

Inclusion and exclusion criteria

Articles were screened for eligibility by A.J.K based on title, abstract, and keywords, and next, if applicable, based on the full text, using Rayyan software (Fig. 1). Inconclusive screening decisions were resolved through consultation with K.R.J. and A.L.B.

FIG. 1.

FIG. 1.

Overview of the number of articles identified and included throughout the several phases of the screening process, depicted according to the PRISMA guidelines.

Articles (e.g., original research, editorials, opinion articles, letters) published in English in a peer-reviewed academic journal in the period January 01, 2008 to October 05, 2021, which discussed an ethical implication of the development or application of TE for regenerative purposes, were eligible for inclusion. Also included were articles that discussed as their main topic ethical or societal implications of RM more generally, provided that these implications were applicable to TE as well.

Excluded were book chapters, conference articles, and news articles, in addition to articles on RM that discussed SC transplantation/research or GT instead of TE. Also excluded were ethical implications related to the source of (stem) cells and the use of embryonic tissues, which have been discussed in depth elsewhere.12–14 Supplementary Data S1 for a more detailed description of the inclusion and exclusion criteria.

As a minimal standard for quality assessment, articles had to be published in a peer-reviewed journal.

Data extraction and analysis

Full texts of the included articles were analyzed systematically using a data extraction sheet (Supplementary Data S1) (A.J.K.). For each article, information was extracted about publication type, publication year, and about the topic and aim, substantiated by relevant quotes. Moreover, one-sentence descriptions of the relevant ethical implications mentioned in each article were extracted (referred to as an implication mention), substantiated by one or more quotes. Quote lengths diverged from one sentence up to page-length. Additional quotes were extracted to preserve contextual information relevant to the ethical implications where necessary.

A thematic analysis was conducted to synthesize the data, using data synthesis sheets (Supplementary Data S1).15 Implication mentions were compared across articles, and combined into implication types based on close similarity (compare “reason mention” and “reason type” as described by Strech and Sofaer)16 (A.J.K.). Implication types were then thematized using a coding tree of four hierarchical levels (i.e., with primary, secondary, and tertiary sub themes under each main theme) (A.J.K.). Each implication type was assigned to one sub theme. If more than one sub theme was applicable, the most suitable one was selected. The formulation of the implication types and the coding tree was then evaluated by all authors and optimized through an iterative process of discussion and adaptation until agreement on a meaningful thematization was reached between all authors (A.J.K., K.R.J., M.C.V., and A.L.B.).

Results

The database search resulted in the identification of 2451 unique articles. Of these, 237 articles were considered eligible and were included in this review (Fig. 1).

Characteristics of included articles

The included articles encompassed reviews in natural sciences (n = 61/237) and humanities (n = 19/237), original research in natural sciences (n = 13/237) and humanities (n = 56/237, of which were 8 empirical and 48 theoretical studies), perspectives (n = 68/237), and other publication types (n = 20/237, of which were 10 workgroup and project reports, 5 interviews, 3 methods articles, 1 case report, and 1 book review) (see Supplementary Data S2). The articles discussed TE for regenerative purposes as a main topic (n = 115/237, including 21 on bioprinted implants and 4 on organoid transplantation) or subtopic (n = 65/237, including as a main topic: 9 on Advanced Therapy Medicinal Products (ATMP), 8 on SC transplantation, 6 on RM, and 6 on in vitro TE), or discussed RM, broadly yet in a manner relevant to TE, as their main topic (n = 51/237) or subtopic (n = 6/237) (Supplementary Data S2). The articles were published in 2008 (n = 10), 2009 (n = 17), 2010 (n = 13), 2011(n = 9), 2012 (n = 10), 2013 (n = 18), 2014 (n = 22), 2015 (n = 33), 2016 (n = 16), 2017 (n = 23), 2018 (n = 14), 2019 (n = 24), 2020 (n = 21), and 2021 (n = 18).

Synthesis of the ethical implications of TE for regenerative purposes

A range of ethical implications of TE for regenerative purposes was identified in the included literature. These were categorized into 10 main themes. The first seven themes cluster phases of TE development from bench to bedside:

  • (1)

    animal welfare

  • (2)

    handling human tissue

  • (3)

    informed consent

  • (4)

    therapeutic potential

  • (5)

    risk and safety

  • (6)

    clinical translation

  • (7)

    societal impact.

The remaining three themes group overarching ethical domains for safeguarding responsible development:

  • (8)

    scientific integrity

  • (9)

    regulation

  • (10)

    patient and public involvement.

The ethical implications (as “implication types”) identified in the literature are discussed below per main theme and sub theme, and are also summarized in Tables 1–10. Supplementary Data S3 for one-sentence descriptions of the “implication mentions” per article corresponding to each aggregated “implication type”.

Table 1.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Animal Welfare

Animal welfare
For tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Animal experimentation … animal welfare should be balanced with scientific need and reliability of animal models 17–29
… in vitro, ex vivo and in silico models should be applied to (partially) replace the use of animals 30–48
… methods should be applied to reduce the number of animals per experiment 49–53
… the use of non-human primates or companion animals should be avoided, or experiments should be refined to minimize suffering 18,27,50,54–58
Animal-derived TE components … animal-free TE scaffold and culture components are preferred 22,38,59–69

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Table 2.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Handling Human Tissue

Handling human tissue
Tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Tissue collection, storage, and use … requires tissue collection methods that are not harmful to the donor 17,22
… requires consent of (the family of) the donor for the collection, storage, and use of their tissues 17,19,20,22,23,58,70–76
… requires that donors be fully informed on future use and possible financial interests, before collection, storage, and use of their tissues 7,77–86
… requires protection of donor privacy when storing and using their tissues and data 7,17,19,22,23,58,73,75,77,78,80,82,86–90
… raises the question under what conditions private, public, or hybrid model biobanks are acceptable 73,74,84,85,88,91–96
… should be composed of non-human-derived scaffold components 72,97–100
Ownership and commodification … raises the question who has ownership over tissue engineered products 7,17,22,23,75,79,88,101–104
… raises the question if tissue engineered products should be patentable 17,73,74,82,101,105–109
… reinforces commercialization of human tissues, which is at odds with altruistic donation and risks donor exploitation 71–74,80,84,88,93,110–115

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Table 3.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Informed Consent

Informed consent
For tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Challenges to informed consent … consent might be challenged by the participant's capacity to consent 75,116
… informed consent might be challenged by therapeutic misconception 19,23,37,74,115,117–119
… the voluntariness of consent might be challenged by pressure from family or researchers 22,120
… informed consent for innovative therapies might be challenged by positive bias of the physician 119,121–123
… informed consent is challenged by the inherent uncertainty involved in risk assessment 20,22,23,71,81,88,104,112,118,119,124,125
Conditions for informed consent … well-considered informed consent requires that patients receive relevant information, e.g., on risk-benefit ratio, cell source, and alternative treatment options 37,74,119,126–128
… well-considered informed consent requires that patients receive support and information in understandable language 17,119,129–131

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Table 4.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Therapeutic Potential

Therapeutic potential
Tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Therapeutic promise … has the potential to provide a cure for diseases where currently non is available or where side effects are high 75,82,106,116,132–135
… may provide an alternative to donor organ transplantation, removing the need for immunosuppression and solving organ shortages 17,23,60,74,75,78,79,81,88,89,101–103,112,117,125,128,133,136–152
… is excisable, which is a benefit compared to other RM strategies 81,125
Objectives for wellbeing … should aim to improve the quality of life of patients 20,153

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Table 5.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Risk and Safety

Risk and safety
Tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Safety risks … bears risks related to the presence of living cells, such as immunogenicity, mutagenesis, and tumorigenicity 22,23,70,75,79,81,86,88,89,101,124,125,141,154–156
… bears risks related to the scaffold components, such as contamination, immunogenicity, and cytotoxicity 22,23,72,79,81,116,124,125,155–158
… bears safety risks related to the complexity of TE constructs in terms of variability, dynamic interaction, and irreversibility 3,25,70,75,78–81,102,115,119,122,124,132
… involves inherent uncertainties related to the complexity of TE and to the limited predictive value of animal models 3,23,71,80,88,104,153,159–161
Safety measures … requires that risks are minimized and are proportional to benefits to the individual and society 3,23,37,73,126,131,132,158,161,162
… requires appropriate preclinical testing before first-in-human trials 26,29,37,120,124,126,129,131,154,161,163
… requires long-term follow-up of trial participants 37,58,120,124,154,164–166
… requires that TE implants are detectable in case of emergency situations 102
… requires that safety measures are not unnecessarily burdensome 131,132,165,167

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Table 6.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Clinical Translation

Clinical translation
For tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Clinical trial design … randomized controlled trials might be challenging if no standard of care is available and if sham surgery would be required as a control 131,161,167–173
… first-in-human clinical trials should include patients with mid- or end-stage disease 3,19,23,37,70,73,74,81,124,161,168,174,175
… first-in-human clinical trials should include efficacy endpoints and capture both objective and subjective outcomes 21,37,70,73,75,81,125,126,131,155,156,160,164,167,168,176
… registries are needed to collect long-term safety and efficacy data 3,17,37,74,86,101,117,120,126,131,167,177
… clinical trials should be designed carefully, and results should be published transparently, to avoid bias and promote reproducibility 71,74,125,127,163,164,178,179
Innovative therapy pathway … innovative therapy as alternative to clinical trials may promote patient access to experimental treatments 23,37,74,122,180
… innovative therapy as alternative to clinical trials is at tension with the need to protect patients from risks and with the need for evidence collection 7,19,121,123,131,166,178,181–183
Unproven commercial interventions … unproven interventions expose patients to the risk of physical harm and financial exploitation 19,74,120,122,123,156,178,180,184–186
… researchers and physicians should protect patients against the risks of unproven interventions 7,18,120,183,187,188

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Table 7.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Societal Impact

Societal impact
Tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Distributive justice … could exacerbate local and global socioeconomic health disparities 17,19,22,23,70,75,79,88,102,104,107,140,158,180,189–195
… should be developed to be cost effective to improve accessibility 25,37,74,89,90,112,124,155,166,170,196–198
… poses a value trade-off between the future fruits of innovation and current patient demands 71,74,88,104,137,146,158,169,199–202
… should be reimbursed based on clinical effectiveness, costs, and patient preferences 19,102,123,154,167,178,182
Impact on health care and markets … might provide an alternative to ethically contentious health care practices like organ donation, xenotransplantation, and gestational motherhood 133,140,149,203–205
… could help ending illegal organ trade 79,90,140,141,206
Biosecurity … bears risks of bioterrorism related to unregulated home-use 17,81,107,125,190,207
Longevity and enhancement … raises the question whether we should pursue longevity or accept ageing as a normal part of life 88,103,110,192,193,208–211
… might change social acceptability of life organ donation and unhealthy lifestyles 17,88,101,108,192,212
… raises opportunities and concerns related to the possibility of enhancement 20,23,70,75,88,108,110,112,158,180,190,213,214
Human experience and identity … raises presents the body as malleable and challenges our conception of what it means to be human 17,88,101,104,110,115,180,189,210,215–223
… blurs the boundaries between the natural and the artificial 17,88,140,223–225

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Table 8.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Scientific Integrity

Scientific integrity
For tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Responsible conduct … researchers and media should refrain from overstated promises to prevent hype 23,85,88,112,137,140,146,156,169,178,180,191,194,226
… a benign research environment and better oversight mechanisms are needed to prevent fraud 129,169,227,228
… scientists should (be trained to) be aware of the societal and ethical implications of their work 3,25,37,74,102,107,168,192,229–231
Transdisciplinary collaboration … successful development co-depends on transdisciplinary and international collaborations between scientists, clinicians, regulators, and industry 37,92,102,106,121,125,131,137,177,182,232–236
… commercial pressure might lead to premature testing and commercialization, so that government sponsored trials are preferable 17,20,67,80,89,106,115,118,131,162,168,174,177,178,180,194,237
… responsible development is fostered by involvement of ethics and social science scholars in the innovation process 17,38,106,140,181,238,239

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Table 9.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Regulation

Regulation
For tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Guidelines and oversight … adequate regulatory frameworks should be established and be harmonized internationally 70,103,120,154,157,178,180,181,234,240
… uniform guidelines and oversight for the use of human tissues should be established 84,85,93,128,136,162,165,241
… uniform safety, quality and efficacy standards, and controls for TE products and processes should be established 23,25,75,106,108,120,124,126,127,132,171,174,179,195,242–245
… regulation of innovative therapies offered outside clinical trials is needed 19,122,153
… regulation should foster a balance between safety requirements and speed of innovation, to promote patient access 3,20,22,74,80,102,111,135,144,246
… clarification or revision of the current regulatory classification of TE products is needed 17,78,79,81,89,101,126,217,224
Liability … it should be determined who is responsible for quality control and maintenance and who should be held liable if complications arise 20,75,83,101,102,112,121,126,217

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Table 10.

Overview of the Ethical Implications of Tissue Engineering for Regenerative Purposes, as Identified in Relation to Patient and Public Involvement

Patient and public involvement
For tissue engineering for regenerative purposes …
 
Sub theme Ethical implications References
Public engagement … the public should be educated on the opportunities, hurdles, and ethical aspects 23,82,88,108,131,147,158,180,181,185,245,247
… patients and the public should be consulted to include their values and needs 20,71,74,82,88,107,175,181,195,230,233,237,248,249
… (bio)art may pose critical questions and provide new spaces for public dialog 38,215,222,250,251

The ethical implications are presented as aggregated “implication types”, categorized into sub themes. See Supplementary Data S3 for one-sentence descriptions of every ethical implication as mentioned in each relevant article (i.e., “implication mentions”).

Animal welfare

The literature discusses ethical implications related to animal welfare that can be subdivided between conditions for preclinical testing on animals and concerns related to animal-derived TE components (Table 1 and Supplementary Data S3).

Animal experimentation

Animal welfare should be balanced with the scientific need and expected human benefit of animal models.17,18 The design should therefore take the predictive value of a particular animal model for humans into account19–24 and the experiments should be performed in accordance with the 3R principles (replace, reduce, and refine).25–29

To replace animals for biomaterial testing or to study regenerative processes, the use of computational modeling30 or in vitro models31–33 are advocated. These include SCs,34 organoids,35 decellularized tissue,36 and organs-on-a-chip.37,38 Ex vivo tissue culture systems39–44 and the chorioallantoic membrane assay45–48 are proposed as means of partial replacement. To reduce the number of animals per experiment, continuous monitoring of each animal,49–51 testing of multiple biomaterials per animal,52 and skipping small animal models in favor of one large animal model53 are suggested. Last, a preference is expressed not to use companion animals,18,27,50,54,55 such as dogs, or primates56 as large animal models, and, if primates have to be used, to refine the experiments by minimizing the suffering caused.57,58

Animal-derived TE components

Animal-derived decellularized scaffolds and culture medium components might raise religious and moral objections related to xenotransplantation and animal welfare, especially among religious or vegan patients.22,59–62 Autologous,63 allogeneic,64–66 or plant-based components67–69 are proposed as alternatives.

Handling human tissue

The literature identifies ethical considerations regarding how to handle human tissues. These can be grouped in two sub themes: first, the collection, storage, and use of cells and tissues from human donors for research and clinical applications, and second, ownership over and commodification of donated and engineered tissues (Table 2 and Supplementary Data S3).

Tissue collection, storage, and use

Tissues should be collected through non-invasive methods that do not cause harm or pain to the donor.17,22 Consent of the donor or their family is required for collection and storage of tissues,22,23,58,70–72 which should be explicit19,73 and voluntary.74,75 Some suggest that consent should be specific to the intended use17 while others argue that broad consent suffices.20,76 As a prerequisite for consent, donors should be provided relevant information, including information on present and foreseeable use of the collected tissues,7,77–81 on the need to forego financial rights over the tissue,82 and on policies outlining the return of findings83,84 and the possibility to withdraw.85,86

To safeguard the confidentiality and privacy of the donor of collected and stored samples,19,22,58,80,82,86,87 some suggest that sample anonymization is desirable,7,17,77 while others point out that this would limit clinical value73 and the possibility of return of findings23 and of dynamic (ongoing) consent.88 Particularly, safeguarding privacy and confidentiality of the (medical) data and digital models required to create personalized 3D bioprinted tissues is warranted.75,78,89,90

A dispute exists whether private (commercial) biobanking is acceptable91 or that public banking should be preferred74 for tissue storage. Specifically, private banking is said to be based on a (false) promise of yet-to-be-developed RM treatments,88,92 which could erode public trust.85,93 Yet public banks would fail in providing a supply that is inclusive to all.84 A hybrid model of public-private biobanking is therefore advocated that allows privately stored tissues to become publicly available, increasing the publicly available supplies94–96 and accelerating research.73

Finally, moral and religious objections could be raised to the use of (decellularized) human tissues, especially cadaveric tissues, as scaffold components. Synthetic scaffolds,97 animal-derived scaffolds,72,98 or (decellularized) tissues from living donors99,100 would therefore be preferred.

Ownership and commodification

The question is raised which parties may claim ownership over donated7,22,23 and engineered17,75,79,88,90,101–104 tissues: the tissue donor, the patient receiving the implant, or the parties involved in producing the organ? Moreover, it is debated whether and under what conditions intellectual property rights over TE, for example, by patenting the technology or the product, is desirable. While patenting could foster innovation,105 ensure quality control, and prevent misuse,17,101 it might also hinder open science and research82,106 and equal access to the fruits of the technology.74,107,108 According to some, notwithstanding property interests of other parties, patients should retain a right to control certain uses and access to the organ.73,109 Finally, TE creates a paradoxical and potentially undesirable situation in which altruistically donated tissues are turned into profitable materials for which donors have no right to financial gain.71–74,80,84,93,110 This appears at odds with human dignity111 and the ban on the commodification of human body parts,112 and might lead to donor exploitation or coercion.88,113–115

Informed consent

The literature mentions informed consent as a necessary precondition for several phases of TE development: for tissue donation (as discussed under ‘Tissue collection, storage, and use’), participation in clinical trials, and clinical application, whether as innovative therapy, compassionate use, or routine treatment. A distinction can be made between challenges and conditions for obtaining valid, informed consent (Table 3 and Supplementary Data S3).

Challenges to consent

Consent might be challenged if the patient's capacity to consent is impaired, for example, in emergency situations116 or when treated for brain conditions.75 Second, patients' hope or desperation for a cure might lead them to misunderstand the risky and experimental nature of a TE trial (therapeutic misconception) and affect the validity of their consent.19,23,37,74,115,117–119 Third, patients might feel pressured to participate by family members or researchers.22,120 Fourth, possible positive bias and conflicting financial or professional interest of the physician-researcher involved in an experimental TE treatment might affect the neutrality of the information provided.119,121–123 And last, uncertainty about possible complications due to the novel,81 complex,22,119,124 invasive,20,118 and irreversible125 nature of TE makes it difficult to provide accurate information about the risk-benefit ratio of the intervention.23,71,88,104,112

Conditions for consent

Due to these challenges, medical professionals should provide, and carefully explain, all relevant information, including information on the characteristics of the TE intervention itself, the source of cells and other components, foreseeable risks and benefits, safety measures, alternative treatment options, and a disclosure of interests.37,119,126–128 Yet, even if high quality information is provided, consent should always be accompanied by oversight mechanisms to prevent patients from unacceptable risks.74 Moreover, information should be provided in understandable language,17,129 and may be supported by the use of educational materials119 and consultation of an independent medical professional.130,131

Therapeutic potential

One of the main ethical opportunities and key drivers of TE is its therapeutic potential. The literature elaborately discusses its therapeutic promise, and briefly its objectives related to broader wellbeing (Table 4 and Supplementary Data S3).

Therapeutic promise

TE is proposed to address an unmet clinical need, having the potential to treat diseases for which currently no curative therapy is available75,106,116,132–134 or for which current therapies have risky side effects,135 and to improve the quality of life of elderly patients.82 Most often, TE is presented as an alternative to donor organ transplantation. As such it would remove the need for immunosuppression,133,136 improve length and quality of life60,74,79,88,103,128,137 and psychosocial outcomes (for face TE),138,139 provide patient-tailored75,81,89,125,140 and/or off-the-shelf available78,141,142 treatment options, and reduce the cost of care.23,143 In the long run it might solve organ shortages,102,117,144–148 thus saving lives and avoiding complexities related to organ donation, including the surgical risks for donors,149,150 the need for (prior or proxy) consent,112,151,152 and the acceptability of directed donation.17,101 Moreover, compared to other RM strategies such as SC injections or GT, cell scaffolding provides TE the benefit of excision if needed, thus allowing partial reversibility.81,125

Objectives for wellbeing

Finally, TE should not merely be aimed at treating disease, but also at improving overall quality of life20 and promoting human flourishing.153

Risk and safety

Notwithstanding the therapeutic potential of TE, the literature discusses several risks of harm for the patients or trial participants, along with safety measures (Table 5 and Supplementary Data S3).

Risks to participant

Risks are associated with different aspects of TE constructs: The presence of living cells poses risks of immunogenicity,70,88,154 mutagenesis (especially for bioprinting),89,101,124,155 undirected differentiation,81,125,156 and teratoma and tumor formation,22,23,75,79,86,141 which are compounded by bioprinting forces. TE components such as scaffolds and bioactive factors pose risks of contamination and introduction of pathogens,22,23,79,124,155,157 potential immunogenicity,72,158 and cytotoxicity (especially of degradation products).81,116,125,156

The complexity of the TE constructs themselves pose risks that are foreseeable but hard to quantify, that is, risks associated with inherent variability of the constructs,78,79,119 unpredictable dynamic interactions with the bodily environment,25,80,124,132 and the irreversibility of the surgical implantation and regeneration process.3,102,115,122 The latter particularly impedes patient's opportunity to withdraw from the trial.70,75,81 In addition to the known risks, TE involves inherent uncertainties, because of the complexity and manufacturing variability of the construct,23,71,159 and because the predictive value of preclinical animal studies is limited.3,153 As a result it is challenging to provide a realistic risk-benefit analysis,88,104,160 and to decide when evidence suffices to start first-in-human trials.80,161

Safety measures

Trials might be justified if adequate safety measures are taken to minimize risks131,132,162 and if the benefits to the individual (compared to available therapies)3,126,161 and society (in terms of scientific value)23,37,73,158 are maximized. The following safety measures are described: To understand the biological mechanisms underlying TE126,154 and to evaluate safety and efficacy of the technology, thorough in vitro163 and in vivo26,129,131,161 preclinical research is required and must be optimized to minimize translational distance.29,37,120,124 To monitor adverse events and the success of regeneration, there is a need for long-term follow-up of trial participants,37,58,154,164–166 and pediatric patients in particular.120,124 Artificial organs should be identifiable in case emergency situations arise.102 However, safety measures should not be unnecessarily burdensome131,132 or hamper patient access.165,167

Clinical translation

The pathway of TE treatments from experimental design toward clinical application can roughly take three routes: through clinical trials (i.e., traditional pathway, aimed at evidence collection for research), as innovative therapy (i.e., non-research pathways aimed at improving patient access, including off-label use, compassionate use, and hospital exemption), or interventions can simply reach the market before evidence of efficacy and safety is complete (i.e., unproven commercial interventions). The literature discusses ethical implications related to each of these pathways (Table 6 and Supplementary Data S3).

Clinical trial design

Several suggestions are provided on how to optimize the design of clinical trials for TE. First, while randomized controlled trials (RCT's) should be used where possible,168,169 this might be ethically challenging, especially if no standard of care is available.167,170–172 Sham surgery may be required as a comparator, but is justifiable only if surgery is minimally invasive and if anticipated social value outweighs harm to the control group.131,161,173 Moreover, adequate and fair participant selection is challenging,19,73 because fewer patients can be enrolled than are eligible,74 little is known about comorbidities,174 and end-stage patients are especially vulnerable to therapeutic misconception.23,37

Still, consensus seems to exist that for high-risk TE, performing first-in-human phase I trials on healthy volunteers or stable patients is unacceptable.3 Patients with mid- or end-stage disease should be included instead,70,81,124,161,168,175 because they might benefit most and risk losing least. Regarding outcome measures, first-in-human trials may involve efficacy endpoints in addition to safety endpoints,73,81 to maximize the scientific value. Efficacy endpoints should include objective outcomes, such as overall survival167 and success of tissue regeneration,126,164 evaluation of which should preferably not rely on invasive biopsies,21,176 and subjective outcomes related to patient wellbeing.37,131,160,168 For personalized (3D bioprinted) implants, heterogeneous outcomes are to be expected and limit the possibility to extrapolate the results.70,75,125,155,156

Finally, curated registries should be set up and used to collect long-term data from clinical trials and follow-up studies regarding safety and efficacy of TE interventions.3,17,37,74,86,101,120,126,167 This would allow to trace adverse events,177 help build a solid evidence-base, and allow to evaluate when the intervention needs no longer be considered experimental.117,131 Moreover, in combination with general non-biased and transparent publishing practices and the use of scientifically valid and rigorous research methods,71,74,125,127,163,164,178,179 this could promote reproducibility and foster translation of TE products to the clinic.

Innovative therapy pathway

Because the use of traditional clinical trials for TE is faced by several challenges, alternatives are needed. One option is to allow patients to access experimental RM and TE treatments as innovative therapy outside clinical trials. This would improve patients access74,122,180 and circumvent the need for sham surgery.23,37 However, such access might be at tension with the need to protect patients from considerable risk,7,121,178 and the need to generate conclusive evidence regarding safety and efficacy of the product123,181,182 and could therefore harm public trust. Publishing outcomes and adverse events in open registries might partially overcome these concerns.19,131,166,183

Unproven commercial interventions

Commercial clinics also offer SC-based RM interventions (which could include TE interventions) without sufficient proof of efficacy and safety. Authors warn that such unproven interventions operate outside existing regulations and are based on false promises,123,180,184,185 thus exposing vulnerable patients to the risk of physical harm and financial exploitation,19,74,156,178,186 and harming public trust in the RM field.120,122 Both researchers and physicians are responsible to inform patients and protect them against these risks.7,18,120,183,187,188

Societal impact

The literature discusses several societal impacts of TE and RM development. These include so-called “soft” impacts related to human values, experiences, identity, relations, and perceptions. The literature covers challenges to distributive justice, the impact on health care and market, biosecurity, longevity and enhancement, and human experience and identity (Table 7 and Supplementary Data S3).

Distributive justice

Concern exists that the expected high costs of TE interventions will limit its accessibility. If only those who can afford to pay will benefit in terms of quality and length of life,17,75,140,189–191 this may exacerbate socioeconomic health disparities19,22,23,70,107,158,192,193 both locally102 and globally.79,88,104,180,194,195 However, if, in the long run, the costs of TE will decrease, treatments could become cost effective.89,112,166,196 Efforts should be made toward that end so as to improve accessibility.25,37,74,90,124,155,170,197,198 Additionally, to ensure equal access to TE treatments, reimbursement will be necessary154,182 and reimbursement decisions should be based on evidence of clinical benefit,123 patient preferences,167 and initial and lifetime costs102,167,178 compared to current treatments options.

Moreover, while there is an imperative to fund RM and TE research,199,200 this might draw resources away from existing health care solutions, exposing a value trade-off between the future fruits of innovation and the demand of patients in need today.71,74,88,104 Resources should therefore be allocated based on good science rather than hype137,146,169 and arrangements for benefit-sharing (such as price limits or revenue sharing) should be put in place.158,201,202

Impact on health care and markets

TE might provide an alternative to several ethically contentious health care practices: TE organs might circumvent personal identity issues related to organ donation,203 and naturalness objections against xenotransplantation.140 Transplantable bioartificial uteri might provide an alternative to gestational surrogate motherhood149,204,205 and could possibly contribute to the realization of human ectogenesis on the long run.133 TE organs could also help ending illegal organ trade,79,141,206 provided that regulations are adopted to prevent new black markets in TE organs.90,140

Biosecurity

Concern is raised that TE technology, and particularly 3D bioprinting because of its relative accessibility and affordability, might lead to unregulated home-use, which could facilitate bioterrorism.17,81,107,125,190,207

Longevity and enhancement

TE might help to prolong the average human life span or even “cure aging”, which raises the question whether longevity is socially desirable193,208,209 or that ageing should be viewed as a normal part of life.88,103,192 In particular, an expanded life span might impact the retirement system210 and could decrease solidarity with older generations.110,211 If TE tissues and organs would become widely available, this could also affect social norms: unhealthy and reckless lifestyles might be normalizsed17,88,101,108,192 and support for organ donation might decrease.212 Moreover, TE could be used for cosmetic purposes or potentially to enhance physical or cognitive abilities. While the pursuit of enhancement is deeply embedded in human culture23,75,112 and could help to pursue the good life,88 it raises concerns about potential harm,108 increasing inequalities,70,158 fair resource distribution,20,110 and the legitimate goals of medicine.180,190,213,214

Human experience and identity

TE presents the body as malleable110,189,215 or machine-like.104 This raises the question whether the human body or life itself is something that should be engineered,17,115,180,216 and might challenge our understanding of what it means to be human,210,217,218 and our perception of the self219,220 and of others.101 The engineering ideal driving TE might obscure the existential and embodied experiences of patients.88,221–223 Last, TE organs blur the boundaries between the corporal and the technical,88,224 life and non-life,140,225 and natural and engineered. While its social acceptance might depend on whether it will be perceived as the one or the other,17 TE is best presented as engineered.223

Scientific integrity

The literature identifies issues of scientific integrity related to individual conduct and to transdisciplinary collaboration (Table 8 and Supplementary Data S3).

Responsible conduct

TE, SC and organoid research, and organ bioprinting are surrounded by hype and high public expectations.88,180,194 To prevent disillusionment and promote public trust, researchers, scientific journals, and popular media should set realistic expectations,140,146,169,178,191,226 by making modest claims and refraining from overstated promises.23,85,112,137,156 The TE field has been faced with a serious case of scientific misconduct and research fraud by the thoracic surgeon Paolo Macchiarini. This indicates the need for a more benign research environment and better oversight mechanisms within TE, to promote integrity of conduct and to restore trust in the field.129,169,227,228 Responsible conduct requires that scientists express scientific virtues such as openness, honesty, and accountability74,107 and that they anticipate how their choices affect the final TE product and its beneficiaries.3,37,102 They should therefore be trained to become aware of and be able to engage in discourse on the societal and ethical implications of TE.25,168,192,229–231

Transdisciplinary collaboration

Successful TE development co-depends on transdisciplinary collaboration between engineers, scientists, and clinicians102,137,232 in a globally inclusive manner182,233; adequate training of physician-researchers to gain necessary expertise knowledge and skills to implement TE safely in clinical trials37,121,131,234,235; infrastructures of knowledge exchange106; and mutual collaboration and relationships of trust between academia, industry, and regulators.92,125,177,236

However, industry involvement and commercial pressure might steer the research agenda,106 incentivize selection of trial sites in areas with permissive regulations,177,180,237 and lead to premature clinical testing and commercialization of TE products.20,115,118,174,178,194 To deal with potential conflicts of interest and to maintain trust from physicians and the public, independent oversight mechanisms are needed,80,89,168 full disclosure of interests should be provided by those involved in clinical trials67,131 and trials sponsored by governments rather than industry might be preferred.17,162

Finally, ethics, humanities, and social science scholars should be involved in the innovation process to take ethical, legal, and societal considerations into account, to manage potential conflicts of interest and to promote successful and responsible translation of TE to the clinic.17,38,106,140,181,238,239

Regulation

The need for adequate regulatory frameworks for TE is widely discussed in the literature. The relevant ethical implications can be divided in the need for appropriate guidelines and oversight, and liability issues (Table 9 and Supplementary Data S3).

Need for guidelines and oversight

The regulatory frameworks guiding TE development should be revaluated.103,120,181 Particularly, given regional disparities in TE regulations, many authors call for international harmonization70,154,157,178,240 or, at minimum, agreement on an international code of conduct.180,234 In particular, guidelines and adequate oversight should be established for the collection, storage, and use of human tissues84,93,128,136,162,241 and for ownership rights over such tissues.85,165 Moreover, there is a need to set uniform safety, quality, and efficacy standards for cell culture,120,126 biomaterials,23,25,242,243 manufacturing processes,75,108,124 and TE end products106,171,195,244,245 and to establish ethical oversight127,132 or regulatory controls174,179 to guide risk assessment. Finally, global regulations to guide the application of experimental TE and RM treatments as innovative therapy outside clinical trials are needed.19,122,153

A particular challenge for TE regulation is fostering a balance between the product safety and speed of product development.3,20 While regulations should prevent commercial pressure from impeding good patient care,102 regulations should not be unnecessarily risk-averse, so as not to slow down progress,135,144,246 drive up costs, or impede patient access.74,111 This relates to the question when clinical evidence suffices to introduce TE products into regular clinical practice.22,80 Another regulatory challenge for TE and 3D-bioprinted products is that–because they combine features of drugs, devices, and biologicals and because of their personalized naturethey fall in between most currently existing classificatory categories.78,217 These classifications should therefore be urgently revised to provide clarity.17,79,81,89,101,126,224

Liability

Considering the multidisciplinary nature of TE development, it should be determined who (regulator, engineer, manufacturer, or clinician) is responsible for quality assurance112 and long-time maintenance102 of TE implants and who should be held liable in case serious complications arise.20,75,83,101 This is especially challenging in case a patient decides to withdraw from a trial.126 While one author warns that currently patient protection lags behind because companies can avoid liability for TE product failure too easily,217 another argues that physicians should be better protected from negligence claims if innovative therapies turn out harmful, in order not to stifle innovation.121

Patient and public involvement

The literature shows a general interest in involving patients and the public in a dialog on TE development, to promote public trust and to enable scientific citizenship (Table 10 and Supplementary Data S3). As a start, the public should be educated on the opportunities and hurdles, along with ethical and societal aspects of TE. This requires scientists to communicate clearly about their research, and provide accessible and reliable information sources,23,82,88,108,131,147,158,180,181,245,247 or even set up counseling services to counter misinformation.185

Next, a wide range of stakeholders, including lay people and patients, should be consulted on their values, needs, expectations, and concerns regarding TE development.74,82,88,195,233,248 In particular, such consultation could probe the acceptability of risks,20,237 approaches to deal with uncertainties,71 and fair distribution of health care resources.175,230 Patient advocates are especially well-positioned to articulate patient needs.181,249 One author group points out that to foster truly inclusive deliberation, scientists should be attentive to a wide variety of stakeholders and empower them to make informed and meaningful contributions.107

Finally, (bio)artists are particularly well positioned to explain or visualize scientific work,38 to critically question the TE field, its implications and assumptions215,250 and to provide alternative places for discourse on the intersection of art and bioethics.222,251

Discussion

In this SR of 237 academic peer-reviewed articles, we identified a range of ethical implications of TE for regenerative purposes. These stretch over 10 overarching themes, representing phases of TE development and implementation (animal welfare, handling human tissue, informed consent, therapeutic potential, risk and safety, clinical translation, and societal impact), and safeguards necessary for responsible development (scientific integrity, regulation, and public and patient involvement).

A prior literature review performed in 2008, De Vries et al noted that until then the literature on the ethics of TE and RM mainly focused on considerations relating to the source and collection of SCs in preclinical research and that the ethical evaluation of TE clinical trials lacked short.5 Our SR reveals that, since then, ethical reflection in academic discourse has followed the pace of TE development, shifting focus from preclinical to clinical translation, and providing several concrete recommendations on how this process should take place.

Our SR indicates several characteristics of TE that are specific to the ethical implications of the product. That is, (a) TE is highly innovative23 and has a novel method and aim,3,161 that is, tissue regeneration rather than “mere” restoration of function; (b) TE products are increasingly personalised,23 for example, bioprinted from patient-derived cells after a patient-specific shape; (c) TE products deeply invade161 in and dynamically interact3,126 with the human body and finally disappear88; and (d) TE is multidisciplinary38,119,140 in nature and relies on state-of-the-art technologies such as 3D (bio)printing, iPSCs, and organoid technology. Several ethical implications related to these characteristics are touched upon in this review but remain in need of further ethical reflection.

First, the novel aim and personalized character of TE challenge traditional clinical trial pathways and guidance is needed on how to proceed with clinical translation of TE. The possibility to perform traditional RCTs or to extrapolate the results from one patient to another is limited because suitable comparators are lacking, and TE interventions are patient specific. N-of-1 trials have been proposed as an alternative,252,253 but guidelines are needed on how to reduce risks for individuals while maximizing the scientific value of the outcomes. Alternatively, patients could be provided access to unproven interventions as innovative therapies outside clinical trials when individual benefit can be reasonably expected.23,74 This may be beneficial provided that a learning component, including registration and reporting, is maintained.122,254

Second, TE constructs invade and integrate deeply into the human body, which may have implications for how the technology is perceived and experienced. For example, TE may impact embodiment and perceptions of (un)naturalness. While these so-called “soft” impacts on society are often indirect and intangible, their effect may be significant.4,255 Therefore, we agree with others88,192,256 that more efforts should be made to anticipate and evaluate the soft mid- and long-term impacts of TE and RM on society at large.

Third, the multidisciplinary nature and commercial incentives in the TE field raise questions to whom ownership rights and responsibilities regarding TE products should be ascribed. While this could be approached as a strictly legal question on the patentability of tissue products and processes,105,109 a broader ethical analysis is needed that takes into account how individuals perceive ownership over TE implants. It has been shown that intuitions about ownership over tissues derived from one's own body tend to deviate from ownership rights as laid down in existing regulations.257 Both the value attached to cells and tissue contructs,84,258,259 and the invasive character of TE implants that integrate deeply into the body could and should affect what degree of ownership and control is attributed and to whom. This will have implications for who will gain financially, and how much, and could therefore also impact the accessibility of TE interventions. Broadening the notion of ownership could help justify benefit sharing schemes73,201,202 to improve access and to let patients and the public share in the benefits of TE and RM innovations.

Methodological choices, strengths, and limitations

Because the focus of the TE debate has moved toward settling conditions for responsible development and anticipating future implications, we opted for an SR of “ethical implications” rather than “reasons for/against” as is sometimes done for ethics literature.11,16 We broadly interpreted “ethical implications” to include considerations of a philosophical, existential, artistic, legal, regulatory, and anticipatory nature and questions of governance, public and patient involvement, and (scientific) misconduct.

We restricted the scope of our SR to “TE for regenerative purposes” because of its clinical significance and because we expected that at least some of the ethical implications would be related to the particular characteristics and purpose of TE thus defined. However, demarcating “TE for regenerative purposes” as distinct from other forms of RM was challenging because in the literature different definitions of “TE” and “RM” are used,260 but often not explicated. Moreover, in practice, TE, SC interventions, and GT often overlap or are combined. A detailed list of inclusion and exclusion criteria that was used to guide this demarcation is provided in Supplementary Data S1.

Overall, we present a comprehensive review that covers over two hundred articles, indicating a broad set of ethical considerations related to all phases of TE development, including potential future societal impact. Three possible limitations should be mentioned. First, by excluding book chapters and conference articles we may have missed a small number of ethical implications. Second, both phases of literature screening (title/abstract screening and full text screening) were performed by one reviewer only, a method for which support exists261 but that might be more fallible than traditional double-blind screening. However, as has been argued elsewhere,262 we may have captured all (or most) relevant ethical implications, even if a small number of articles was missed. Third, while we touch upon several regulatory considerations, we did not discuss existing regulatory regimes in detail, as this is outside the scope of our review. However, others have recently compared ATMP and bioprinting regulations across jurisdictions.79,263

Concluding Remarks

Overall, as TE and RM shift toward clinical translation, it raises ethical questions regarding fundamental and clinical research, translation to the patient, and its impact on society at large. To our knowledge, this is the first time that the ethical implications of this field have been reviewed systematically. By gathering existing scholarly work and identifying knowledge gaps, this SR facilitates further research into the ethical and societal implications of TE and RM. Moreover, it will serve as a valuable resource and educational tool for TE and RM scientists, engineers, and clinicians by providing an overview of the ethical considerations relevant to their work. As such, our review will help promote successful and responsible development of new interventions in TE and RM in a manner attentive of the current and future ethical and societal implications encountered along the way. Given its broad and comprehensive scope, it may also serve as a steppingstone for the ethical analysis of other emerging biomedical technologies.

Supplementary Material

Supplemental data
Supp_DataS1.pdf (169KB, pdf)
Supplemental data
Supp_DataS2.pdf (124.1KB, pdf)
Supplemental data
Supp_DataS3.pdf (772.8KB, pdf)

Acknowledgments

We thank Paulien Wiersma of the Utrecht University Library for revising the database search strategy. We would like to thank our colleagues from the Medical Humanities Department for their valuable comments on earlier versions of this work.

Authors' Contributions

All authors contributed to conceptualization and writing of this article.

Disclosure Statement

Annelien Bredenoord is a member of the Dutch Senate. She also serves as a member of the Ethics Committee of the ISSCR and the Ethics Advisory Board of IQVIA. The other authors have no conflicts of interest to declare.

Funding Information

This research was financially supported by the Gravitation Program “Materials Driven Regeneration,” funded by the Netherlands Organization for Scientific Research (024.003.013).

Supplementary Material

Supplementary Data S1

Supplementary Data S2

Supplementary Data S3

References

  • 1. Mason C, Dunnill P. A brief definition of regenerative medicine. Regen Med 2008;3(1):1–5; doi: 10.2217/17460751.3.1.1 [DOI] [PubMed] [Google Scholar]
  • 2. Wissing TB, Bonito V, Bouten CVC, et al. Biomaterial-driven in situ cardiovascular tissue engineering—a multi-disciplinary perspective. NPJ Regen Med 2017;2(1):1–20; doi: 10.1038/s41536-017-0023-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Trommelmans L, Selling J, Dierickx K. Is tissue engineering a new paradigm in medicine? Consequences for the ethical evaluation of tissue engineering research. Med Health Care Philos 2009;12(4):459–467; doi: 10.1007/s11019-009-9192-0 [DOI] [PubMed] [Google Scholar]
  • 4. Jongsma KR, Bredenoord AL. Ethics parallel research: An approach for (early) ethical guidance of biomedical innovation. BMC Med Ethics 2020;21(81):1–9; doi: 10.1186/s12910-020-00524-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. De Vries RBM, Oerlemans AJM, Trommelmans L, et al. Ethical aspects of tissue engineering: A review. Tissue Eng Part B Rev 2008;14(4):367–375; doi: 10.1089/ten.teb.2008.0199 [DOI] [PubMed] [Google Scholar]
  • 6. Kim YS, Smoak MM, Melchiorri AJ, et al. An Overview of the Tissue Engineering Market in the United States from 2011 to 2018. Tissue Eng Part A 2019;25(1–2):1–8; doi: 10.1089/ten.tea.2018.0138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. O'Donnell BT, Ives CJ, Mohiuddin OA, et al. Beyond the present constraints that prevent a wide spread of tissue engineering and regenerative medicine approaches. Front Bioeng Biotechnol 2019;7:1–12; doi: 10.3389/fbioe.2019.00095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Slingerland AS, Smits AIPM, Bouten CVC. Then and now: Hypes and hopes of regenerative medicine. Trends Biotechnol 2013;31(3):121–123; doi: 10.1016/j.tibtech.2012.12.001 [DOI] [PubMed] [Google Scholar]
  • 9. Kahrass H, Borry P, Gastmans C, et al. PRISMA-Ethics – Reporting Guideline for Systematic Reviews on Ethics Literature: Development, explanations and examples. OSF Prepr 2021; doi: 10.31219/osf.io/g5kfb [DOI] [Google Scholar]
  • 10. Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. Ann Intern Med 2009;151(4):264–269; doi: 10.7326/0003-4819-151-4-200908180-00135 [DOI] [PubMed] [Google Scholar]
  • 11. Mertz M, Kahrass H, Strech D. Current state of ethics literature synthesis: A systematic review of reviews. BMC Med 2016;14(1):1–12; doi: 10.1186/s12916-016-0688-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Hug K, Hermerén G. Do we still need human embryonic stem cells for stem cell-based therapies? Epistemic and Ethical Aspects. Stem Cell Rev Reports 2011;7(4):761–774; doi: 10.1007/s12015-011-9257-3 [DOI] [PubMed] [Google Scholar]
  • 13. Zarzeczny A, Caulfield T. Emerging ethical, legal and social issues associated with stem cell research & and the current role of the moral status of the embryo. Stem Cell Rev Reports 2009;5(2):96–101; doi: 10.1007/s12015-009-9062-4 [DOI] [PubMed] [Google Scholar]
  • 14. Power C, Rasko JEJ. Will cell reprogramming resolve the embryonic stem cell controversy? a narrative review. Ann Intern Med 2011;155(2):114–121; doi: 10.7326/0003-4819-155-2-201107190-00007 [DOI] [PubMed] [Google Scholar]
  • 15. Barnett-Page E, Thomas J. Methods for the synthesis of qualitative research: A critical review. BMC Med Res Methodol 2009;9(1):59; doi: 10.1186/1471-2288-9-59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Strech D, Sofaer N. How to write a systematic review of reasons. J Med Ethics 2012;38(2):121–126; doi: 10.1136/medethics-2011-100096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Vijayavenkataraman S, Lu WF, Fuh JYH. 3D bioprinting – An Ethical, Legal and Social Aspects (ELSA) framework. Bioprinting 2016;1:11–21; doi: 10.1016/j.bprint.2016.08.001 [DOI] [Google Scholar]
  • 18. Rozlin AR, Muhammad Aa'zamuddin AR, Norhamiza MS, et al. Tissue engineering of articular cartilage: From bench to bed-side. Tissue Eng Regen Med 2014;12(1):1–11; doi: 10.1007/s13770-014-9044-8 [DOI] [Google Scholar]
  • 19. Lowenthal J, Sugarman J. Ethics and policy issues for stem cell research and pulmonary medicine. Chest 2015;147(3):824–834; doi: 10.1378/chest.14-1696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Nicholson J. Reflections on the ethics of biomaterials science. New Bioeth 2013;19(1):54–63; doi: 10.1179/2050287713z.00000000021 [DOI] [PubMed] [Google Scholar]
  • 21. Sculean A, Nikolidakis D, Nikou G, et al. Biomaterials for promoting periodontal regeneration in human intrabony defects: A systematic review. Periodontol 2000 2015;68(1):182–216; doi: 10.1111/prd.12086 [DOI] [PubMed] [Google Scholar]
  • 22. Oerlemans AJM, Rodrigues CHCML, Verkerk MA, et al. Ethical aspects of soft tissue engineering for congenital birth defects in children-what do experts in the field say? Tissue Eng Part B Rev 2010;16(4):397–403; doi: 10.1089/ten.teb.2009.0666 [DOI] [PubMed] [Google Scholar]
  • 23. Otto IA, Breugem CC, Malda J, et al. Ethical considerations in the translation of regenerative biofabrication technologies into clinic and society. Biofabrication 2016;8(4):042001; doi: 10.1088/1758-5090/8/4/042001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Liguori GR, Jeronimus BF, De Aquinas Liguori TT, et al. Ethical Issues in the Use of Animal Models for Tissue Engineering: Reflections on Legal Aspects, Moral Theory, Three Rs Strategies, and Harm'Benefit Analysis. Tissue Eng Part C Methods 2017;23(12):850–862; doi: 10.1089/ten.tec.2017.0189 [DOI] [PubMed] [Google Scholar]
  • 25. Kashi A, Saha S. Ethics in biomaterials research. J Long Term Eff Med Implants 2009;19(1):19–30; doi: 10.1615/jlongtermeffmedimplants.v19.i1.30 [DOI] [PubMed] [Google Scholar]
  • 26. Levin D. Bench to bedside: Approaches for engineered intestine, esophagus, and colon. Gastroenterol Clin North Am 2019;48(4):607–623; doi: 10.1016/j.gtc.2019.08.012 [DOI] [PubMed] [Google Scholar]
  • 27. Moran CJ, Ramesh A, Brama PAJ, et al. The benefits and limitations of animal models for translational research in cartilage repair. J Exp Orthop 2016;3(1):1–12; doi: 10.1186/s40634-015-0037-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Falcon JM, Karchner JP, Henning EA, et al. Ethics of using animal models as predictors of human response in tissue engineering. Ethics Biol Eng Med 2019;10(1):37–49; doi: 10.1615/EthicsBiologyEngMed.2020033718 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Abbas TO, Elawad A, Pullattayil S, et al. Quality of reporting in preclinical urethral tissue engineering studies: A systematic review to assess adherence to the arrive guidelines. Animals 2021;11(8):2456; doi: 10.3390/ani11082456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Tartarini D, Mele E. Adult stem cell therapies for wound healing: Biomaterials and computational models. Front Bioeng Biotechnol 2016;3:1–7; doi: 10.3389/fbioe.2015.00206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Dikici S, Aldemir Dikici B, Bhaloo SI, et al. Assessment of the Angiogenic Potential of 2-Deoxy-D-Ribose Using a Novel in vitro 3D dynamic model in comparison with established in vitro assays. Front Bioeng Biotechnol 2020;7:1–20; doi: 10.3389/fbioe.2019.00451 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Sauer UG. Animal and non-animal experiments in nanotechnology - The results of a critical literature survey. ALTEX 2009;26(2):109–127; doi: 10.14573/altex.2009.2.109 [DOI] [PubMed] [Google Scholar]
  • 33. Lammers G, Tjabringa GS, Schalkwijk J, et al. A molecularly defined array based on native fibrillar collagen for the assessment of skin tissue engineering biomaterials. Biomaterials 2009;30(31):6213–6220; doi: 10.1016/j.biomaterials.2009.07.050 [DOI] [PubMed] [Google Scholar]
  • 34. Ahmed U, Ahmed R, Masoud MS, et al. Stem cells based in vitro models: Trends and prospects in biomaterials cytotoxicity studies. Biomed Mater 2021;16(4); doi: 10.1088/1748-605X/abe6d8 [DOI] [PubMed] [Google Scholar]
  • 35. Ghareeb AE, Lako M, Steel DH. Coculture techniques for modeling retinal development and disease, and enabling regenerative medicine. Stem Cells Transl Med 2020;9(12):1531–1548; doi: 10.1002/sctm.20-0201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. McCrary MW, Bousalis D, Mobini S, et al. Decellularized tissues as platforms for in vitro modeling of healthy and diseased tissues. Acta Biomater 2020;111:1–19; doi: 10.1016/j.actbio.2020.05.031 [DOI] [PubMed] [Google Scholar]
  • 37. Baker HB, McQuilling JP, King NMP. Ethical considerations in tissue engineering research: Case studies in translation. Methods 2016;99:135–144; doi: 10.1016/j.ymeth.2015.08.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Edwards J, Thomas R, Guilliatt R. Regenerative medicine: From the laboratory looking out. Palgrave Commun 2017;3(1):27; doi: 10.1057/s41599-017-0036-x [DOI] [Google Scholar]
  • 39. Smith EL, Kanczler JM, Oreffo ROC. A new take on an old story: Chick limb organ culture for skeletal niche development and regenerative medicine evaluation. Eur Cells Mater 2013;26:91–106; doi: 10.22203/eCM.v026a07 [DOI] [PubMed] [Google Scholar]
  • 40. Smith EL, Kanczler JM, Gothard D, et al. Evaluation of skeletal tissue repair, Part 1: Assessment of novel growth-factor-releasing hydrogels in an ex vivo chick femur defect model. Acta Biomater 2014;10(10):4186–4196; doi: 10.1016/j.actbio.2014.06.011 [DOI] [PubMed] [Google Scholar]
  • 41. Weightman AP, Pickard MR, Yang Y, et al. An in vitro spinal cord injury model to screen neuroregenerative materials. Biomaterials 2014;35(12):3756–3765; doi: 10.1016/j.biomaterials.2014.01.022 [DOI] [PubMed] [Google Scholar]
  • 42. Peeters M, Van Rijn S, Vergroesen PPA, et al. Bioluminescence-mediated longitudinal monitoring of adipose-derived stem cells in a large mammal ex vivo organ culture. Sci Rep 2015;5:1–12; doi: 10.1038/srep13960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Richardson SM, Hoyland JA, Mobasheri R, et al. Mesenchymal stem cells in regenerative medicine: Opportunities and challenges for articular cartilage and intervertebral disc tissue engineering. J Cell Physiol 2010;222(1):23–32; doi: 10.1002/jcp.21915 [DOI] [PubMed] [Google Scholar]
  • 44. Sloan AJ, Lynch CD. Dental tissue repair: Novel models for tissue regeneration strategies. Open Dent J 2013;6(1):214–219; doi: 10.2174/1874210601206010214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Moreno-Jiménez I, Kanczler JM, Hulsart-Billstrom G, et al. The chorioallantoic membrane assay for biomaterial testing in tissue engineering: A short-term in vivo preclinical model. Tissue Eng Part C Methods 2017;23(12):938–952; doi: 10.1089/ten.tec.2017.0186 [DOI] [PubMed] [Google Scholar]
  • 46. Abubakar AA, Noordin MM, Azmi TI, et al. The use of rats and mice as animal models in ex vivo bone growth and development studies. Bone Jt Res 2016;5(12):610–618; doi: 10.1302/2046-3758.512.BJR-2016-0102.R2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Baiguera S, Macchiarini P, Ribatti D. Chorioallantoic membrane for in vivo investigation of tissue-engineered construct biocompatibility. J Biomed Mater Res Part B Appl Biomater 2012;100 B(5):1425–1434; doi: 10.1002/jbm.b.32653 [DOI] [PubMed] [Google Scholar]
  • 48. Rashidi H, Sottile V. The chick embryo: Hatching a model for contemporary biomedical research. BioEssays 2009;31(4):459–465; doi: 10.1002/bies.200800168 [DOI] [PubMed] [Google Scholar]
  • 49. Yeo DC, Wiraja C, Mantalaris A, et al. Nanosensors for regenerative medicine. J Biomed Nanotechnol 2014;10(10):2722–2746; doi: 10.1166/jbn.2014.1992 [DOI] [PubMed] [Google Scholar]
  • 50. Hahn JA, Witte TS, Arens D, et al. Double-plating of ovine critical sized defects of the tibia: A low morbidity model enabling continuous in vivo monitoring of bone healing. BMC Musculoskelet Disord 2011;12:2–7; doi: 10.1186/1471-2474-12-214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Trachtenberg JE, Vo TN, Mikos AG. Pre-clinical characterization of tissue engineering constructs for bone and cartilage regeneration. Ann Biomed Eng 2015;43(3):681–696; doi: 10.1007/s10439-014-1151-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Oliveira MB, Ribeiro MP, Miguel SP, et al. In vivo high-content evaluation of three-dimensional scaffolds biocompatibility. Tissue Eng Part C Methods 2014;20(11):851–864; doi: 10.1089/ten.tec.2013.0738 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. De Vries RBM, Buma P, Leenaars M, et al. Reducing the number of laboratory animals used in tissue engineering research by restricting the variety of animal models. Articular cartilage tissue engineering as a case study. Tissue Eng Part B Rev 2012;18(6):427–435; doi: 10.1089/ten.teb.2012.0059 [DOI] [PubMed] [Google Scholar]
  • 54. Alvites RD, Branquinho MV, Sousa AC, et al. Small ruminants and its use in regenerative medicine: Recent works and future perspectives. Biology (Basel) 2021;10(3):1–21; doi: 10.3390/biology10030249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Mangione F, Salmon B, EzEldeen M, et al. Characteristics of large animal models for current cell-based oral tissue regeneration. Tissue Eng Part B Rev 2022;28(3):489–505; doi: 10.1089/ten.teb.2020.0384 [DOI] [PubMed] [Google Scholar]
  • 56. Quinn RW. Animal models for bench to bedside translation of regenerative cardiac constructs. Prog Pediatr Cardiol 2013;35(2):91–94; doi: 10.1016/j.ppedcard.2013.08.001 [DOI] [Google Scholar]
  • 57. Baklaushev VP, Durov OV, Kim SV, et al. Development of a motor and somatosensory evoked potentials-guided spinal cord Injury model in non-human primates. J Neurosci Methods 2019;311:200–214; doi: 10.1016/j.jneumeth.2018.10.030 [DOI] [PubMed] [Google Scholar]
  • 58. Afshar L, Aghayan H, Sadighi J, et al. Ethics of research on stem cells and regenerative medicine: Ethical guidelines in the Islamic Republic of Iran. Stem Cell Res Ther 2020;11(396):1–5; doi: 10.1186/s13287-020-01916-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Wellings D. Novel 3D scaffolds for tissue culture, regenerative medicine and wound care. Sp2 2014;20–22 [Google Scholar]
  • 60. Kanwar S, Vijayavenkataraman S. Design of 3D printed scaffolds for bone tissue engineering: A review. Bioprinting 2021;24:e00167; doi: 10.1016/j.bprint.2021.e00167 [DOI] [Google Scholar]
  • 61. Almarzouqi F, Rennekampff HO, Almarzouki M, et al. Porcine-derived biomaterials in tissue engineering and reconstructive surgery: Considerations and alternatives in Muslim patients. J Tissue Eng Regen Med 2019;13(2):253–260; doi: 10.1002/term.2788 [DOI] [PubMed] [Google Scholar]
  • 62. Munirah S, Zainul Ibrahim Z, Rozlin AR, et al. Exploring the Islamic perspective on tissue engineering principles and practice. Glob J Al-Thaqafah 2014;4(2):29–40; doi: 10.7187/gjat642014.04.02 [DOI] [Google Scholar]
  • 63. Offner D, de Grado GF, Meisels I, et al. Bone grafts, bone substitutes and regenerative medicine acceptance for the management of bone defects among french population: Issues about ethics, religion or fear? Cell Med 2019;11:215517901985766; doi: 10.1177/2155179019857661 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Hou T, Li Z, Luo F, et al. A composite demineralized bone matrix - Self assembling peptide scaffold for enhancing cell and growth factor activity in bone marrow. Biomaterials 2014;35(22):5689–5699; doi: 10.1016/j.biomaterials.2014.03.079 [DOI] [PubMed] [Google Scholar]
  • 65. Seo J pil, Tsuzuki N, Haneda S, et al. Comparison of allogeneic platelet lysate and fetal bovine serum for in vitro expansion of equine bone marrow-derived mesenchymal stem cells. Res Vet Sci 2013;95(2):693–698; doi: 10.1016/j.rvsc.2013.04.024 [DOI] [PubMed] [Google Scholar]
  • 66. Tekkatte C, Gunasingh GP, Cherian KM, et al. “Humanized” stem cell culture techniques: The animal serum controversy. Stem Cells Int 2011;2011:504723; doi: 10.4061/2011/504723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Langridge B, Griffin M, Butler PE. Regenerative medicine for skeletal muscle loss: A review of current tissue engineering approaches. J Mater Sci Mater Med 2021;32(1):15; doi: 10.1007/s10856-020-06476-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Zhu Y, Zhang Q, Wang S, et al. Current advances in the development of decellularized plant extracellular matrix. Front Bioeng Biotechnol 2021;9:712262; doi: 10.3389/fbioe.2021.712262 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Contessi Negrini N, Toffoletto N, Farè S, et al. Plant tissues as 3D natural scaffolds for adipose, bone and tendon tissue regeneration. Front Bioeng Biotechnol 2020;8:1–15; doi: 10.3389/fbioe.2020.00723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Ruiz-alonso S, Villate-beitia I, Gallego I, et al. Current insights into 3D bioprinting: An advanced approach for eye tissue regeneration. Pharmaceutics 2021;13(3):1–28; doi: 10.3390/pharmaceutics13030308 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Hermerén G. The ethics of regenerative medicine. Biol Futur 2021;72(2):113–118; doi: 10.1007/s42977-021-00075-3 [DOI] [PubMed] [Google Scholar]
  • 72. Lee E, Milan A, Urbani L, et al. Decellularized material as scaffolds for tissue engineering studies in long gap esophageal atresia. Expert Opin Biol Ther 2017;17(5):573–584; doi: 10.1080/14712598.2017.1308482 [DOI] [PubMed] [Google Scholar]
  • 73. Boers SN, Van Delden JJM, Clevers H, et al. Organoid biobanking: Identifying the ethics. EMBO Rep 2016;17(7):938–941; doi: 10.15252/embr.201642613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Cossu G, Birchall M, Brownc T, et al. Lancet commission: Stem cells and regenerative medicine. Lancet 2018;391(10123):883–910; doi: 10.1016/S0140-6736(17)31366-1 [DOI] [PubMed] [Google Scholar]
  • 75. Kirillova A, Bushev S, Abubakirov A, et al. Bioethical and legal issues in 3D bioprinting. Int J Bioprinting 2020;6(3):1–10; doi: 10.18063/IJB.V6I3.272 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Hempel C, Lomax G, Peckman S. Broad consent in biobanking. Nat Biotechnol 2012;30(9):826; doi: 10.1038/nbt.2349 [DOI] [PubMed] [Google Scholar]
  • 77. Oerlemans AJM, Van Den Berg PP, Van Leeuwen E, et al. Ethical issues regarding the donation and source of cells for tissue engineering: A European focus group study. Tissue Eng Part B Rev 2011;17(4):229–234; doi: 10.1089/ten.teb.2010.0683 [DOI] [PubMed] [Google Scholar]
  • 78. Li P, Faulkner A. 3D Bioprinting Regulations: A UK/EU Perspective. Eur J Risk Regul 2017;8(2):441–447; doi: 10.1017/err.2017.19 [DOI] [Google Scholar]
  • 79. Sekar MP, Budharaju H, Zennifer A, et al. Current standards and ethical landscape of engineered tissues—3D bioprinting perspective. J Tissue Eng 2021;12: 20417314211027677; doi: 10.1177/20417314211027677 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Lu L, Arbit HM, Herrick JL, et al. Tissue engineered constructs: Perspectives on clinical translation. Ann Biomed Eng 2015;43(3):796–804; doi: 10.1007/s10439-015-1280-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Gilbert F, O'Connell CD, Mladenovska T, et al. Print Me an Organ? ethical and regulatory issues emerging from 3D bioprinting in medicine. Sci Eng Ethics 2018;24(1):73–91; doi: 10.1007/s11948-017-9874-6 [DOI] [PubMed] [Google Scholar]
  • 82. Horch RE, Popescu LM, Vacanti C, et al. Ethical issues in cellular and molecular medicine and tissue engineering. J Cell Mol Med 2008;12(5B):1785–1793; doi: 10.1111/j.1582-4934.2008.00460.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Lowdell MW, Thomas A. The expanding role of the clinical haematologist in the new world of advanced therapy medicinal products. Br J Haematol 2017;176(1):9–15; doi: 10.1111/bjh.14384 [DOI] [PubMed] [Google Scholar]
  • 84. Trommelmans L, Selling J, Dierickx K. The importance of the values attached to cells for a good informed consent procedure in cell donation for tissue engineering purposes. Cell Tissue Bank 2009;10(4):293–299; doi: 10.1007/s10561-009-9123-6 [DOI] [PubMed] [Google Scholar]
  • 85. Master Z, Crowley AP, Smith C, et al. Stem cell preservation for regenerative therapies: Ethical and governance considerations for the health care sector. NPJ Regen Med 2020;5(1):23; doi: 10.1038/s41536-020-00108-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Gómez-Barrena E, Rosset P, Müller I, et al. Bone regeneration: Stem cell therapies and clinical studies in orthopaedics and traumatology. J Cell Mol Med 2011;15(6):1266–1286; doi: 10.1111/j.1582-4934.2011.01265.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Reinke S, Dienelt A, Blankenstein A, et al. Qualifying stem cell sources: How to overcome potential pitfalls in regenerative medicine? J Tissue Eng Regen Med 2016;10(1):3–10; doi: 10.1002/term.1923 [DOI] [PubMed] [Google Scholar]
  • 88. Oerlemans AJM, van Hoek MEC, van Leeuwen E, et al. Towards a richer debate on tissue engineering: A Consideration on the Basis of NEST-Ethics. Sci Eng Ethics 2013;19(3):963–981; doi: 10.1007/s11948-012-9419-y [DOI] [PubMed] [Google Scholar]
  • 89. Ravnic DJ, Leberfinger AN, Koduru SV, et al. Transplantation of bioprinted tissues and organs: Technical and clinical challenges and future perspectives. Ann Surg 2017;266(1):48–58; doi: 10.1097/SLA.0000000000002141 [DOI] [PubMed] [Google Scholar]
  • 90. Singh S, Choudhury D, Yu F, et al. In situ bioprinting – Bioprinting from benchside to bedside? Acta Biomater 2020;101:14–25; doi: 10.1016/j.actbio.2019.08.045 [DOI] [PubMed] [Google Scholar]
  • 91. Roberts DE. Umbilical cord blood banking: Public good or private benefit? Med J Aust 2008;189(10):599; doi: 10.5694/j.1326-5377.2008.tb02197.x [DOI] [PubMed] [Google Scholar]
  • 92. Martin P, Brown N, Turner A. Capitalizing hope: The commercial development of umbilical cord blood stem cell banking. New Genet Soc 2008;27(2):127–143; doi: 10.1080/14636770802077074 [DOI] [Google Scholar]
  • 93. Pirnay JP, Baudoux E, Cornu O, et al. Access to human tissues for research and product development: From EU regulation to alarming legal developments in Belgium. EMBO Rep 2015;16(5):557–562; doi: 10.15252/embr.201540070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Stewart CL, Aparicio LC, Kerridge IH. Ethical and legal issues raised by cord blood banking - The challenges of the new bioeconomy. Med J Aust 2013;199(4):290–292; doi: 10.5694/mja12.11668 [DOI] [PubMed] [Google Scholar]
  • 95. McGuckin CP, Forraz N.. Umbilical cord blood stem cells - An ethical source for regenerative medicine. Med Law 2008;27(1):147–165. [PubMed] [Google Scholar]
  • 96. Ballen KK, Verter F, Kurtzberg J. Umbilical cord blood donation: Public or private? Bone Marrow Transplant 2015;50(10):1271–1278; doi: 10.1038/bmt.2015.124 [DOI] [PubMed] [Google Scholar]
  • 97. Hikita A, Chung U Il, Hoshi K, et al. Bone regenerative medicine in oral and maxillofacial region using a three-dimensional printer. Tissue Eng Part A 2017;23(11–12):515–521; doi: 10.1089/ten.tea.2016.0543 [DOI] [PubMed] [Google Scholar]
  • 98. Crowe CS, Chiou G, McGoldrick R, et al. In vitro characteristics of porcine tendon hydrogel for tendon regeneration. Ann Plast Surg 2016;77(1):47–53; doi: 10.1097/SAP.0000000000000361 [DOI] [PubMed] [Google Scholar]
  • 99. Muhammad Aa'zamuddin AR, Sha'ban M, Azharuddin NSM, et al. A Survey on Relevant Malaysian Fatwā about cadaver concerning tissue engineering research. Pertanika J Soc Sci Humanit 2020;28(2):1451–1466 [Google Scholar]
  • 100. Muhammad Aa'zamuddin AR, Nur Syamimi M, Abdurezak A, et al. Relevant Local Fatwā on the issues of using human tissues in articular cartilage tissue engineering experimentation. IIUM Med J Malaysia 2021;20(1):127–135; doi: 10.31436/IMJM.V20I1.1763 [DOI] [Google Scholar]
  • 101. Vijayavenkataraman S. A perspective on bioprinting ethics. Artif Organs 2016;40(11):1033–1038; doi: 10.1111/aor.12873 [DOI] [PubMed] [Google Scholar]
  • 102. Hutchison K, Sparrow R. What pacemakers can teach us about the ethics of maintaining artificial organs. Hastings Cent Rep 2016;46(6):14–24; doi: 10.1002/hast.644 [DOI] [PubMed] [Google Scholar]
  • 103. Jeyaraj R, Natasha G, Kirby G, et al. Vascularisation in regenerative therapeutics and surgery. Mater Sci Eng C 2015;54:225–238; doi: 10.1016/j.msec.2015.05.045 [DOI] [PubMed] [Google Scholar]
  • 104. Trommelmans L. The challenge of regenerative medicine. Hastings Cent Rep 2010;40(6):24–26; doi: 10.1002/j.1552-146X.2010.tb00074.x [DOI] [PubMed] [Google Scholar]
  • 105. Furman E. The dynamic state of patents in regenerative medicine. Tissue Eng Regen Med 2013;10(5):230–233; doi: 10.1007/s13770-013-1082-0 [DOI] [Google Scholar]
  • 106. Lavoie M. The role of social scientists in accelerating innovation in regenerative medicine. Rev Policy Res 2011;28(6):613–630; doi: 10.1111/j.1541-1338.2011.00525.x [DOI] [Google Scholar]
  • 107. Sample M, Boulicault M, Allen C, et al. Multi-cellular engineered living systems: Building a community around responsible research on emergence. Biofabrication 2019;11(4):043001; doi: 10.1088/1758-5090/ab268c [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Jamieson C, Keenan P, Kirkwood D, et al. A review of recent advances in 3D bioprinting with an eye on future regenerative therapies in veterinary medicine. Front Vet Sci 2021;7:1–13; doi: 10.3389/fvets.2020.584193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Harbaugh JT. Do you own your 3D bioprinted body? Analyzing property issues at the intersection of digital information and biology. Am J Law Med 2015;41(1):167–189; doi: 10.1177/0098858815591512 [DOI] [PubMed] [Google Scholar]
  • 110. Lafontaine C. Regenerative medicine's immortal body: From the fight against ageing to the extension of longevity. Body Soc 2009;15(4):53–71; doi: 10.1177/1357034X09347223 [DOI] [Google Scholar]
  • 111. Pirnay JP, Vanderkelen A, De Vos D, et al. Business oriented EU human cell and tissue product legislation will adversely impact member states' health care systems. Cell Tissue Bank 2013;14(4):525–560; doi: 10.1007/s10561-013-9397-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Patuzzo S, Goracci G, Gasperini L, et al. 3D bioprinting technology: Scientific aspects and ethical issues. Sci Eng Ethics 2018;24(2):335–348; doi: 10.1007/s11948-017-9918-y [DOI] [PubMed] [Google Scholar]
  • 113. Haddad C. Services in the self: Embodied labor and the global bioeconomy. Life Sci Soc Policy 2015;11(9); doi: 10.1186/s40504-015-0027-x [DOI] [Google Scholar]
  • 114. Tyagi P, Dhindsa M.. Tissue engineering and its applications in dentistry. Indian J Dent Res 2009;20(2):222–226. [DOI] [PubMed] [Google Scholar]
  • 115. Leabu M. A framework for assessment and management of ethical risks related to stem cell use in tissue engineering. Ethics Biol Eng Med 2011;2(4):333–345; doi: 10.1615/ethicsbiologyengmed.2012006025 [DOI] [Google Scholar]
  • 116. Das S, Carnicer-Lombarte A, Fawcett JW, et al. Bio-inspired nano tools for neuroscience. Prog Neurobiol 2016;142:1–22; doi: 10.1016/j.pneurobio.2016.04.008 [DOI] [PubMed] [Google Scholar]
  • 117. Lim ML, Jungebluth P, Ajalloueian F, et al. Whole organ and tissue reconstruction in thoracic regenerative surgery. Mayo Clin Proc 2013;88(10):1151–1166; doi: 10.1016/j.mayocp.2013.03.011 [DOI] [PubMed] [Google Scholar]
  • 118. Madonna R, Van Laake LW, Davidson SM, et al. Position Paper of the European Society of Cardiology Working Group Cellular Biology of the Heart: Cell-based therapies for myocardial repair and regeneration in ischemic heart disease and heart failure. Eur Heart J 2016;37(23):1789–1798; doi: 10.1093/eurheartj/ehw113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Trommelmans L, Selling J, Dierickx K. Informing participants in clinical trials with ex vivo human tissue-engineered products: What to tell and how to tell it? J Tissue Eng Regen Med 2008;2(4):236–241; doi: 10.1002/term.82 [DOI] [PubMed] [Google Scholar]
  • 120. Best TM, Caplan A, Coleman M, et al. Not missing the future: A call to action for investigating the role of regenerative medicine therapies in pediatric/adolescent sports injuries. Curr Sports Med Rep 2017;16(3):202–210; doi: 10.1249/JSR.0000000000000357 [DOI] [PubMed] [Google Scholar]
  • 121. Keren-Paz T, El Haj AJ. Liability versus innovation: The legal case for regenerative medicine. Tissue Eng Part A 2014;20(19–20):2555–2560; doi: 10.1089/ten.tea.2013.0324 [DOI] [PubMed] [Google Scholar]
  • 122. Hyun I. Allowing innovative stem cell-based therapies outside of clinical trials: Ethical and policy challenges. J Law Med Ethics 2010;38(2):277–285; doi: 10.1111/j.1748-720X.2010.00488.x [DOI] [PubMed] [Google Scholar]
  • 123. Lysaght T. Accelerating regenerative medicine: The Japanese experiment in ethics and regulation. Regen Med 2017;12(6):657–668; doi: 10.2217/rme-2017-0038 [DOI] [PubMed] [Google Scholar]
  • 124. Oerlemans AJM, Feitz WFJ, Van Leeuwen E, et al. Regenerative urology clinical trials: An ethical assessment of road blocks and solutions. Tissue Eng Part B Rev 2013;19(1):41–47; doi: 10.1089/ten.teb.2012.0136 [DOI] [PubMed] [Google Scholar]
  • 125. Jovic TH, Combellack EJ, Jessop ZM, et al. 3D Bioprinting and the Future of Surgery. Front Surg 2020;7:1–10; doi: 10.3389/fsurg.2020.609836 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Trommelmans L, Selling J, Dierickx K. Ethical reflections on clinical trials with human tissue engineered products. J Med Ethics 2008;34(9):e1; doi: 10.1136/jme.2007.022913 [DOI] [PubMed] [Google Scholar]
  • 127. Trommelmans L, Selling J, Dierickx K. An exploratory survey on the views of european tissue engineers concerning the ethical issues of tissue engineering research. Tissue Eng Part B Rev 2009;15(3):241–247; doi: 10.1089/ten.teb.2008.0647 [DOI] [PubMed] [Google Scholar]
  • 128. Parent B, Gelb B, Latham S, et al. The ethics of testing and research of manufactured organs on brain-dead/recently deceased subjects. J Med Ethics 2020;46(3):199–204; doi: 10.1136/medethics-2019-105674 [DOI] [PubMed] [Google Scholar]
  • 129. Arnason G. Regulating clinical innovation: Trachea transplants and tissue engineering. Am J Bioeth 2019;19(6):32–34; doi: 10.1080/15265161.2019.1602179 [DOI] [PubMed] [Google Scholar]
  • 130. O'Keefe RJ, Tuan RS, Lane NE, et al. American Society for bone and mineral research-orthopaedic research society joint task force report on cell-based therapies. J Bone Miner Res 2020;35(1):3–17; doi: 10.1002/jbmr.3839 [DOI] [PubMed] [Google Scholar]
  • 131. Taylor DA, Caplan AL, Macchiarini P. Ethics of bioengineering organs and tissues. Expert Opin Biol Ther 2014;14(7):879–882; doi: 10.1517/14712598.2014.915308 [DOI] [PubMed] [Google Scholar]
  • 132. The Committee for Advanced Therapies (CAT). Challenges with advanced therapy medicinal products and how to meet them. Nat Rev Drug Discov 2010;9(3):195–201; doi: 10.1038/nrd3052 [DOI] [PubMed] [Google Scholar]
  • 133. Huh Y, Kim YY, Ku SY. Perspective of bioartificial uterus as gynecological regenerative medicine. Tissue Eng Regen Med 2012;9(5):233–239; doi: 10.1007/s13770-012-0360-6 [DOI] [Google Scholar]
  • 134. Muhammad Aa'zamuddin AR, Hashi AA, Sha'ban M. An overview of the application of Maqasid Al-Shariah into cartilage tissue engineering. Int Med J Malaysia 2016;17(Specialissue1):151–156; doi: 10.31436/imjm.v17i1.1013 [DOI] [Google Scholar]
  • 135. Gaissmaier C, Koh JL, Weise K, et al. Future perspectives of articular cartilage repair. Injury 2008;39(1 Suppl):114–120; doi: 10.1016/j.injury.2008.01.033 [DOI] [PubMed] [Google Scholar]
  • 136. Haag JC, Jungebluth P, MacChiarini P. Tracheal replacement for primary tracheal cancer. Curr Opin Otolaryngol Head Neck Surg 2013;21(2):171–177; doi: 10.1097/MOO.0b013e32835e212b [DOI] [PubMed] [Google Scholar]
  • 137. Oerlemans AJM, Van Hoek MEC, Van Leeuwen E, et al. Hype and expectations in tissue engineering. Regen Med 2014;9(1):113–122; doi: 10.2217/rme.13.89 [DOI] [PubMed] [Google Scholar]
  • 138. Simonacci F, Toni R, Raposio E.. Facial transplantation. An update of results and perspectives from tissue engineering. Ann Ital Chir 2017;88:352–359. [PubMed] [Google Scholar]
  • 139. Zou Q, Tian X, Luo S, et al. Agarose composite hydrogel and PVA sacrificial materials for bioprinting large-scale, personalized face-like with nutrient networks. Carbohydr Polym 2021;269:118222; doi: 10.1016/j.carbpol.2021.118222 [DOI] [PubMed] [Google Scholar]
  • 140. Vermeulen N, Haddow G, Seymour T, et al. 3D bioprint me: A socioethical view of bioprinting human organs and tissues. J Med Ethics 2017;43(9):618–624; doi: 10.1136/medethics-2015-103347 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Daar AS. The future of replacement and restorative therapies: From organ transplantation to regenerative medicine. Transplant Proc 2013;45(10):3450–3452; doi: 10.1016/j.transproceed.2013.09.011 [DOI] [PubMed] [Google Scholar]
  • 142. Jank BJ, Xiong L, Moser PT, et al. Engineered composite tissue as a bioartificial limb graft. Biomaterials 2015;61:246–256; doi: 10.1016/j.biomaterials.2015.04.051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Siemionow M. Vascularized composite allotransplantation: A new concept in musculoskeletal regeneration. J Mater Sci Mater Med 2015;26(12):266; doi: 10.1007/s10856-015-5601-5 [DOI] [PubMed] [Google Scholar]
  • 144. Feil G, Daum L, Amend B, et al. From tissue engineering to regenerative medicine in urology - The potential and the pitfalls. Adv Drug Deliv Rev 2011;63(4):375–378; doi: 10.1016/j.addr.2010.12.003 [DOI] [PubMed] [Google Scholar]
  • 145. Rajab TK, Tchantchaleishvili V. Can tissue engineering produce bioartificial organs for transplantation? Artif Organs 2019;43(6):536–541; doi: 10.1111/aor.13443 [DOI] [PubMed] [Google Scholar]
  • 146. Xinaris C. Organoids for replacement therapy: Expectations, limitations and reality. Curr Opin Organ Transplant 2019;24(5):555–561; doi: 10.1097/MOT.0000000000000680 [DOI] [PubMed] [Google Scholar]
  • 147. Brockbank KGM, Swaja RE, Wueste DE, et al. Resolving the shortage of organs for transplantation: Ethics, science, and technology. J S C Med Assoc 2010;106(6):209–212. [PubMed] [Google Scholar]
  • 148. Vijayavenkataraman S, Yan W, Lu WF, et al. 3D bioprinting of tissues and organs for regenerative medicine. Adv Drug Deliv Rev 2018;132:296–332; doi: 10.1016/j.addr.2018.07.004 [DOI] [PubMed] [Google Scholar]
  • 149. Hellström M, Bandstein S, Brännström M. Uterine tissue engineering and the future of uterus transplantation. Ann Biomed Eng 2017;45(7):1718–1730; doi: 10.1007/s10439-016-1776-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Hammond-Browning N. UK criteria for uterus transplantation: A review. BJOG An Int J Obstet Gynaecol 2019;126(11):1320–1326; doi: 10.1111/1471-0528.15844 [DOI] [PubMed] [Google Scholar]
  • 151. Zaami S, Di Luca A, Marinelli E. Advancements in uterus transplant: New scenarios and future implications. Eur Rev Med Pharmacol Sci 2019;23(2):892–902; doi: 10.26355/eurrev_201901_16904 [DOI] [PubMed] [Google Scholar]
  • 152. Swierstra T, van de Bovenkamp H, Trappenburg M. Forging a fit between technology and morality: The Dutch debate on organ transplants. Technol Soc 2010;32(1):55–64; doi: 10.1016/j.techsoc.2010.01.001 [DOI] [Google Scholar]
  • 153. Ventura-Juncá P. Science and ethics: Bridge to the future for regenerative medicine. Int J Stem Cells 2011;4(2):79–84; doi: 10.15283/ijsc.2011.4.2.79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Jacques E, Suuronen EJ. The progression of regenerative medicine and its impact on therapy translation. Clin Transl Sci 2020;13(3):440–450; doi: 10.1111/cts.12736 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Gupta N, Agarwal S. Three-Dimensional Bioprinting: Role in craniomaxillary surgery ethics and future. J Craniofac Surg 2020;31(4):1114–1116; doi: 10.1097/SCS.0000000000006553 [DOI] [PubMed] [Google Scholar]
  • 156. Gilbert F, Viaña JNM, O'Connell CD, et al. Enthusiastic portrayal of 3D bioprinting in the media: Ethical side effects. Bioethics 2018;32(2):94–102; doi: 10.1111/bioe.12414 [DOI] [PubMed] [Google Scholar]
  • 157. Chung E. Stem-cell-based therapy in the field of urology: A review of stem cell basic science, clinical applications and future directions in the treatment of various sexual and urinary conditions. Expert Opin Biol Ther 2015;15(11):1623–1632; doi: 10.1517/14712598.2015.1075504 [DOI] [PubMed] [Google Scholar]
  • 158. Allon I, Ben-Yehudah A, Dekel R, et al. Ethical issues in nanomedicine: Tempest in a teapot? Med Health Care Philos 2017;20(1):3–11; doi: 10.1007/s11019-016-9720-7 [DOI] [PubMed] [Google Scholar]
  • 159. Niemansburg SL, Habets MGJL, Van Delden JJM, et al. Disclosure of risks and uncertainties are especially vital in light of regenerative medicine. Am J Bioeth 2014;14(4):14–16; doi: 10.1080/15265161.2014.889244 [DOI] [PubMed] [Google Scholar]
  • 160. Hogle LF, Das A. The social production of evidence: Regenerative medicine and the 21st Century Cures Act. Regen Med 2017;12(6):581–586; doi: 10.2217/rme-2017-0058 [DOI] [PubMed] [Google Scholar]
  • 161. Niemansburg SL, Van Delden JJM, Dhert WJA, et al. Regenerative medicine interventions for orthopedic disorders: Ethical issues in the translation into patients. Regen Med 2013;8(1):65–73; doi: 10.2217/rme.12.112 [DOI] [PubMed] [Google Scholar]
  • 162. Mathur A, Fernández-Avilés F, Dimmeler S, et al. The consensus of the Task Force of the European Society of Cardiology concerning the clinical investigation of the use of autologous adult stem cells for the treatment of acute myocardial infarction and heart failure: Update 2016. Eur Heart J 2017;38(39):2930–2935; doi: 10.1093/eurheartj/ehw640 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Attico E, Sceberras V, Pellegrini G. Approaches for effective clinical application of stem cell transplantation. Curr Transplant Reports 2018;5(3):244–250; doi: 10.1007/s40472-018-0202-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Taylor DA, Sampaio LC, Gobin A. Building new hearts: A review of trends in cardiac tissue engineering. Am J Transplant 2014;14(11):2448–2459; doi: 10.1111/ajt.12939 [DOI] [PubMed] [Google Scholar]
  • 165. Schneider CK. Advanced therapies: Critical points and perspectives. Pharm Policy Law 2010;12(1–2):25–28; doi: 10.3233/PPL-2010-0260 [DOI] [Google Scholar]
  • 166. Marenah M, Li J, Kumar A, et al. Quality assurance and adverse event management in regenerative medicine for knee osteoarthritis: Current concepts. J Clin Orthop Trauma 2019;10(1):53–58; doi: 10.1016/j.jcot.2018.09.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Jönsson B, Hampson G, Michaels J, et al. Advanced therapy medicinal products and health technology assessment principles and practices for value-based and sustainable healthcare. Eur J Health Econ 2019;20(3):427–438; doi: 10.1007/s10198-018-1007-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Niemansburg SL, Van Delden JJM, Öner FC, et al. Ethical Implications of Regenerative Medicine in Orthopedics: An Empirical Study with Surgeons and Scientists in the Field. Spine J 2014;14(6):1029–1035; doi: 10.1016/j.spinee.2013.10.016 [DOI] [PubMed] [Google Scholar]
  • 169. Brooks M. Stem cell research: Time for a dose of realism. BMJ 2017;356:1–3; doi: 10.1136/bmj.j443 [DOI] [PubMed] [Google Scholar]
  • 170. Trommelmans L, Dierickx K. Standard of care in clinical research with human tissue engineered products (HTEPs). Am J Bioeth 2009;9(3):44–49; doi: 10.1080/15265160802668962 [DOI] [PubMed] [Google Scholar]
  • 171. Hughes B. Christian K. Schneider. Nat Rev Drug Discov 2009;8:270; doi: 10.1038/nrd2864 [DOI] [PubMed] [Google Scholar]
  • 172. Goula A, Gkioka V, Michalopoulos E, et al. Advanced therapy medicinal products challenges and perspectives in regenerative medicine. J Clin Med Res 2020;12(12):780–786; doi: 10.14740/jocmr3964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. De Windt TS, Niemansburg SL, Vonk LA, et al. Ethics in musculoskeletal regenerative medicine; guidance in choosing the appropriate comparator in clinical trials. Osteoarthr Cartil 2019;27(1):34–40; doi: 10.1016/j.joca.2018.08.022 [DOI] [PubMed] [Google Scholar]
  • 174. Arjmand B, Abedi M, Arabi M, et al. Regenerative medicine for the treatment of ischemic heart disease; status and future perspectives. Front Cell Dev Biol 2021;9:1–19; doi: 10.3389/fcell.2021.704903 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Niemansburg SL, Habets MGJL, Dhert WJA, et al. Participant selection for preventive Regenerative Medicine trials: Ethical challenges of selecting individuals at risk. J Med Ethics 2015;41(11):914–916; doi: 10.1136/medethics-2014-102625 [DOI] [PubMed] [Google Scholar]
  • 176. Duarte-Silva M, Guerra-Pinto F, Camelo-Barbosa N, et al. Integration and vascular ingrowth of a collagen meniscal implant: A case report. Malaysian Orthop J 2019;13(2):38–41; doi: 10.5704/MOJ.1907.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Hildebrandt M. Horses for courses: An approach to the qualification of clinical trial sites and investigators in ATMPs. Drug Discov Today 2020;25(2):265–268; doi: 10.1016/j.drudis.2019.10.003 [DOI] [PubMed] [Google Scholar]
  • 178. Kleiderman E, Boily A, Hasilo C, et al. Overcoming barriers to facilitate the regulation of multi-centre regenerative medicine clinical trials. Stem Cell Res Ther 2018;9(1):1–9; doi: 10.1186/s13287-018-1055-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Dohan Ehrenfest DM, Piattelli A, Sammartino G, et al. New Biomaterials and Regenerative Medicine Strategies in Periodontology, Oral Surgery, Esthetic and Implant Dentistry 2018. Biomed Res Int 2019;2019:10–13; doi: 10.1155/2019/1363581 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Chan S. Research translation and emerging health technologies: Synthetic biology and beyond. Health Care Anal 2018;26(4):310–325; doi: 10.1007/s10728-016-0334-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Illes J, Sipp D, Kleiderman E, et al. A blueprint for the next generation of ELSI research, training, and outreach in regenerative medicine. NPJ Regen Med 2017;2(1):21; doi: 10.1038/s41536-017-0026-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Fears R, Akutsu H, Alentajan-Aleta LT, et al. Inclusivity and diversity: Integrating international perspectives on stem cell challenges and potential. Stem Cell Reports 2021;16(8):1847–1852; doi: 10.1016/j.stemcr.2021.07.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Marks PW, Hahn S. Identifying the risks of unproven regenerative medicine therapies. JAMA 2020;324(3):241–242; doi: 10.1001/jama.2020.9375 [DOI] [PubMed] [Google Scholar]
  • 184. Sharma R. Stem cells and tissue engineering in medical practice: Ethical and regulatory policies. Curr Drug Targets 2018;20(4):388–398; doi: 10.2174/1389450119666180831095830 [DOI] [PubMed] [Google Scholar]
  • 185. Smith C, Martin-Lillie C, Higano JD, et al. Challenging misinformation and engaging patients: Characterizing a regenerative medicine consult service. Regen Med 2020;15:1427–1440; doi: 10.2217/rme-2020-0018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Pirnay JP, Vanderkelen A, Zizi M, et al. Human cells and tissues: The need for a global ethical framework. Bull World Health Organ 2010;88(11):870–872; doi: 10.2471/BLT.09.074542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Ryan AL, Ikonomou L, Atarod S, et al. Stem cells, cell therapies, and bioengineering in lung biology and diseases 2017. Am J Respir Cell Mol Biol 2019;61(4):429–439; doi: 10.1165/rcmb.2019-0286ST [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Bauchet L, Lonjon N, Perrin FE, et al. Strategies for spinal cord repair after injury: A review of the literature and information. Ann Phys Rehabil Med 2009;52(4):330–351; doi: 10.1016/j.annrmp.2008.10.004 [DOI] [PubMed] [Google Scholar]
  • 189. Rimmer M. Who owns your body? Patricia Piccinini and the future of bioethics. J Law Med 2013;21(2):370–378. [PubMed] [Google Scholar]
  • 190. Wolinsky H. Printing organs cell-by-cell. EMBO Rep 2014;15(8):836–838; doi: 10.15252/embr.201439207 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Realising the potential of stem-cell research in Europe. Lancet 2013;382(9905):1609; doi: 10.1016/S0140-6736(13)62352-1 [DOI] [PubMed] [Google Scholar]
  • 192. Niemansburg SL, Tempels TH, Dhert WJ, et al. Societal impacts of regenerative medicine: Reflections on the views of orthopedic professionals. Regen Med 2015;10(1):17–24; doi: 10.2217/rme.14.69 [DOI] [PubMed] [Google Scholar]
  • 193. Abeer S. Future medicine: Nanomedicine. J Int Med Sci Acad 2012;25(3):187–192. [Google Scholar]
  • 194. Ripamonti U, Duarte R, Ferretti C. Re-evaluating the induction of bone formation in primates. Biomaterials 2014;35(35):9407–9422; doi: 10.1016/j.biomaterials.2014.07.053 [DOI] [PubMed] [Google Scholar]
  • 195. Lucke JC, Herbert D, Partridge B, et al. Anticipating the use of life extension technologies. EMBO Rep 2010;11(5):334–338; doi: 10.1038/embor.2010.48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Hench LL. The future of bioactive ceramics. J Mater Sci Mater Med 2015;26(2):86; doi: 10.1007/s10856-015-5425-3 [DOI] [PubMed] [Google Scholar]
  • 197. Vemuganti GK, Sangwan VS. Interview: Affordability at the cutting edge: Stem cell therapy for ocular surface reconstruction. Regen Med 2010;5(3):337–340; doi: 10.2217/RME.10.29 [DOI] [PubMed] [Google Scholar]
  • 198. Madeddu P. Cell therapy for the treatment of heart disease: Renovation work on the broken heart is still in progress. Free Radic Biol Med 2021;164:206–222; doi: 10.1016/j.freeradbiomed.2020.12.444 [DOI] [PubMed] [Google Scholar]
  • 199. Malloy C, Wubbenhorst MC, Lee TS. The perinatal revolution. Issues Law Med 2019;34(1):15–41. [PubMed] [Google Scholar]
  • 200. Mhashilkan A, Atala A. Effective Bio-economic approaches for stem cell therapy and regenerative medicine. Curr Stem Cell Res Ther 2012;7(1):1; doi: 10.2174/157488812798483430 [DOI] [PubMed] [Google Scholar]
  • 201. Chapman AR. Patenting in the Public Interest: The California Institute for Regenerative Medicine Model. Am J Bioeth 2018;18(12):61–63; doi: 10.1080/15265161.2018.1531165 [DOI] [Google Scholar]
  • 202. Gardner J. Distributive justice and regenerative medicine. Regen Med 2017;12(7):865–874; doi: 10.2217/rme-2017-0036 [DOI] [PubMed] [Google Scholar]
  • 203. Tamari T. Skin Matters: An Interview with Marc Lafrance. Theory Cult Soc 2019;36(7–8):273–291; doi: 10.1177/0263276419862853 [DOI] [Google Scholar]
  • 204. Miyazaki K, Maruyama T. Partial regeneration and reconstruction of the rat uterus through recellularization of a decellularized uterine matrix. Biomaterials 2014;35(31):8791–8800; doi: 10.1016/j.biomaterials.2014.06.052 [DOI] [PubMed] [Google Scholar]
  • 205. Li X, Wang Y, Ma R, et al. Reconstruction of functional uterine tissues through recellularizing the decellularized rat uterine scaffolds by MSCs in vivo and in vitro. Biomed Mater 2021;16(3):035023; doi: 10.1088/1748-605X/abd116 [DOI] [PubMed] [Google Scholar]
  • 206. Daadi MM, Steinberg GK. Manufacturing neurons from human embryonic stem cells: Biological and regulatory aspects to develop a safe cellular product for stroke cell therapy. Regen Med 2009;4(2):251–263; doi: 10.2217/17460751.4.2.251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Rinaldi T, Colotti G. Use of organoids in medicinal chemistry: Challenges on ethics and biosecurity. Future Med Chem 2019;11(10):1087–1090; doi: 10.4155/fmc-2018-0341 [DOI] [PubMed] [Google Scholar]
  • 208. De Magalhães JP. The scientific quest for lasting youth: Prospects for curing aging. Rejuvenation Res 2014;17(5):458–467; doi: 10.1089/rej.2014.1580 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Zealley B, De Grey ADNJ. Strategies for engineered negligible senescence. Gerontology 2013;59(2):183–189; doi: 10.1159/000342197 [DOI] [PubMed] [Google Scholar]
  • 210. Satava RM. Advanced technologies and the future of medicine and surgery. Yonsei Med J 2008;49(6):873–878; doi: 10.3349/ymj.2008.49.6.873 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Lafontaine C. The postmortal condition: From the biomedical deconstruction of death to the extension of longevity. Sci Cult (Lond) 2009;18(3):297–312; doi: 10.1080/09505430903123008 [DOI] [Google Scholar]
  • 212. Rhodes R. Bioethics: Looking forward and looking back. Am J Bioeth 2013;13(1):13–16; doi: 10.1080/15265161.2013.747318 [DOI] [PubMed] [Google Scholar]
  • 213. Poirot-Mazères I. Legal aspects of the risks raised by nanotechnologies in the field of medicine. J Int Bioethique 2011;22(1):99–118; doi: 10.3917/jib.221.0099 [DOI] [PubMed] [Google Scholar]
  • 214. Djordjevic ML. Penile enhancement surgery. JDDG J der Dtsch Dermatologischen Gesellschaft 2010;8(9):645–647; doi: 10.1111/j.1610-0387.2010.07518.x [DOI] [PubMed] [Google Scholar]
  • 215. Catts O, Zurr I. Growing for different ends. Int J Biochem Cell Biol 2014;56:20–29; doi: 10.1016/j.biocel.2014.09.025 [DOI] [PubMed] [Google Scholar]
  • 216. Maienschein J. Controlling life: From jacques loeb to regenerative medicine. J Hist Biol 2009;42(2):215–230; doi: 10.1007/s10739-009-9180-5 [DOI] [PubMed] [Google Scholar]
  • 217. Parry B. The social life of “Scaffolds”: Examining human rights in regenerative medicine. Sci Technol Hum Values 2018;43(1):95–120; doi: 10.1177/0162243917735179 [DOI] [Google Scholar]
  • 218. Sharp LA. Bioengineered bodies and the moral imagination. Lancet 2009;374(9694):970–971; doi: 10.1016/S0140-6736(09)61653-6 [DOI] [PubMed] [Google Scholar]
  • 219. Horton R. Exhibition: Beauty, the body, and identity. Lancet 2015;385(9977):1499–1500; doi: 10.1016/S0140-6736(15)60741-3 [DOI] [Google Scholar]
  • 220. Moreno J, Gearhart J, Zoloth L, et al. Managing cell and human identity. Science 2017;356(6334):139–140; doi: 10.1126/science.aan2763 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. Devisch I, Vanheule S. Singularity and medicine: Is there a place for heteronomy in medical ethics? J Eval Clin Pract 2014;20(6):965–969; doi: 10.1111/jep.12110 [DOI] [PubMed] [Google Scholar]
  • 222. Couture V, Béisle-Pipon JC, Cloutier M, et al. Merging arts and bioethics: An interdisciplinary experiment in cultural and scientific mediation. Bioethics 2017;31(8):616–630; doi: 10.1111/bioe.12391 [DOI] [PubMed] [Google Scholar]
  • 223. Derksen MHG, Horstman K. Engineering flesh: Towards an ethics of lived integrity. Med Health Care Philos 2008;11(3):269–283; doi: 10.1007/s11019-007-9115-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Fisch M. Regenerating bodies. Sci Technol Hum Values 2018;43(1):121–128; doi: 10.1177/0162243917737364 [DOI] [Google Scholar]
  • 225. Shafer A. “A spark of being”: Frankenstein and anesthesiology. Can J Anesth 2019;66(9):1022–1025; doi: 10.1007/s12630-019-01376-x [DOI] [PubMed] [Google Scholar]
  • 226. Mason C, Manzotti E. Induced pluripotent stem cells: An emerging technology platform and the gartner hype cycle. Regen Med 2009;4(3):329–331; doi: 10.2217/rme.09.20 [DOI] [PubMed] [Google Scholar]
  • 227. Hallonsten O. Corporate culture has no place in academia. Nature 2016;538(7623):7; doi: 10.1038/538007a [DOI] [PubMed] [Google Scholar]
  • 228. Paolo Macchiarini is not guilty of scientific misconduct. Lancet 2015;386(9997):932; doi: 10.1016/S0140-6736(15)00118-X [DOI] [PubMed] [Google Scholar]
  • 229. Merryman WD. Development of a tissue engineered heart valve for pediatrics: A case study in bioengineering ethics. Sci Eng Ethics 2008;14(1):93–101; doi: 10.1007/s11948-008-9053-x [DOI] [PubMed] [Google Scholar]
  • 230. Shineha R, Inoue Y, Ikka T, et al. A comparative analysis of attitudes on communication toward stem cell research and regenerative medicine between the public and the scientific community. Stem Cells Transl Med 2018;7(2):251–257; doi: 10.1002/sctm.17-0184 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Raman R, Bashir R. Biomimicry, Biofabrication, and biohybrid systems: The emergence and evolution of biological design. Adv Healthc Mater 2017;6(20):1–20; doi: 10.1002/adhm.201700496 [DOI] [PubMed] [Google Scholar]
  • 232. Dobrzański LA, Dobrzańska-Danikiewicz AD, Dobrzański LB. Effect of biomedical materials in the implementation of a long and healthy life policy. Processes 2021;9(5):865; doi: 10.3390/pr9050865 [DOI] [Google Scholar]
  • 233. Acero L. Framing regenerative medicine: Culturally specific stories of an emerging technoscience. BioSocieties ; 2021; doi: 10.1057/s41292-021-00236-6 [DOI] [Google Scholar]
  • 234. Epelman I, Murray PE, Garcia-Godoy F, et al. A practitioner survey of opinions toward regenerative endodontics. J Endod 2009;35(9):1204–1210; doi: 10.1016/j.joen.2009.04.059 [DOI] [PubMed] [Google Scholar]
  • 235. Wu AY. The ethical and regulatory-compliant training and practice of regenerative surgery in the United States and Abroad. Stem Cells Dev 2013;22(Suppl 1):54–58; doi: 10.1089/scd.2013.0303 [DOI] [PubMed] [Google Scholar]
  • 236. Gugatschka M, Ohno S, Saxena A, et al. Regenerative medicine of the larynx. where are we today? A review. J Voice 2012;26(5):670.e7–e670.e13; doi: 10.1016/j.jvoice.2012.03.009 [DOI] [PubMed] [Google Scholar]
  • 237. Sleeboom-Faulkner M. Regulatory brokerage: Competitive advantage and regulation in the field of regenerative medicine. Soc Stud Sci 2019;49(3):355–380; doi: 10.1177/0306312719850628 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Garreta E, Kamm RD, Chuva de Sousa Lopes SM, et al. Rethinking organoid technology through bioengineering. Nat Mater 2021;20(2):145–155; doi: 10.1038/s41563-020-00804-4 [DOI] [PubMed] [Google Scholar]
  • 239. Garcia A, Monlezun DJ. Global convergence on the bioethics of surgical implants. Biomed Res Int 2015;2015:853125; doi: 10.1155/2015/853125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Davis NF, Cunnane EM, Mulvihill JJ, et al. The role of stem cells for reconstructing the lower urinary tracts. Curr Stem Cell Res Ther 2018;13(6):458–465; doi: 10.2174/1574888x13666180426113907 [DOI] [PubMed] [Google Scholar]
  • 241. Muhammad Aa'zamuddin AR, Zawawi M, Shaban M, et al. Legal aspects of articular cartilage tissue engineering experimentation: A review on malaysian acts, regulations and guidelines. Malaysian J Med Health Sci 2020;16(3):272–284. [Google Scholar]
  • 242. Moreno-Vega AI, Gómez-Quintero T, Nuñez-Anita RE, et al. Polymeric and ceramic nanoparticles in biomedical applications. J Nanotechnol 2012;2012: 936041; doi: 10.1155/2012/936041 [DOI] [Google Scholar]
  • 243. Romagnoli C. Adipose mesenchymal stem cells in the field of bone tissue engineering. World J Stem Cells 2014;6(2):144; doi: 10.4252/wjsc.v6.i2.144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Okolie O, Stachurek I, Kandasubramanian B, et al. 3d printing for hip implant applications: A review. Polymers (Basel) 2020;12(11):1–29; doi: 10.3390/polym12112682 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245. Bauer H, Heller M, Fink M, et al. Social and legal frame conditions for 3D (and) bioprinting in medicine. Int J Comput Dent 2016;19(4):293–299. [PubMed] [Google Scholar]
  • 246. Keller GS. Biologic treatments are our future, but a more regulated one. Facial Plast Surg Clin North Am 2018;26(4):ix–xi; doi: 10.1016/j.fsc.2018.06.012 [DOI] [PubMed] [Google Scholar]
  • 247. Strech D, Hirschberg I, Meyer A, et al. Ethics Literacy and “Ethics University”: Two intertwined models for public involvement and empowerment in bioethics. Front Public Heal 2016;3:1–9; doi: 10.3389/fpubh.2015.00287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Zoeller K. Science and the lay perspective: Lay people's involvement in assessing tissue engineering. Tissue Eng Part A 2014;20(19–20):2561–2566; doi: 10.1089/ten.tea.2013.0292 [DOI] [PubMed] [Google Scholar]
  • 249. Fernandez A. The making of an advocate. Regen Med 2012;7(6 Suppl):114–116; doi: 10.2217/rme.12.89 [DOI] [PubMed] [Google Scholar]
  • 250. Yetisen AK, Davis J, Coskun AF, et al. Bioart. Trends Biotechnol 2015;33(12):724–734; doi: 10.1016/j.tibtech.2015.09.011 [DOI] [PubMed] [Google Scholar]
  • 251. Holmberg T, Ideland M. Imagination laboratory: Making sense of bio-objects in contemporary genetic art. Soc Rev 2016;64(3):447–467; doi: 10.1111/1467-954X.12387 [DOI] [Google Scholar]
  • 252. Schork NJ. Time for one-person trials. Nature 2015;520(7549):609–611; doi: 10.1038/520609a [DOI] [PubMed] [Google Scholar]
  • 253. Kane PB, Bittlinger M, Kimmelman J. Individualized therapy trials: Navigating patient care, research goals and ethics. Nat Med 2021;27(10):1679–1686; doi: 10.1038/s41591-021-01519-y [DOI] [PubMed] [Google Scholar]
  • 254. Chan S. Current and emerging global themes in the bioethics of regenerative medicine: The tangled web of stem cell translation. Regen Med 2017;12(7):839–851; doi: 10.2217/rme-2017-0065 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Swierstra T. Identifying the normative challenges posed by technology's “soft” impacts. Etikk Praksis 2015;9(1):5–20; doi: 10.5324/eip.v9i1.1838 [DOI] [Google Scholar]
  • 256. Assen LS, Jongsma KR, Isasi R, et al. Recognizing the ethical implications of stem cell research: A call for broadening the scope. Stem Cell Reports 2021;16(7):1656–1661; doi: 10.1016/j.stemcr.2021.05.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Sipp D. Identity and ownership issues in the regulation of autologous cells. Regen Med 2017;12(7):827–838; doi: 10.2217/rme-2017-0063 [DOI] [PubMed] [Google Scholar]
  • 258. Kool EM, Bos AME, van der Graaf R, et al. Ethics of oocyte banking for thirdparty assisted reproduction: A systematic review. Hum Reprod Update 2018;24(5):615–635; doi: 10.1093/humupd/dmy016 [DOI] [PubMed] [Google Scholar]
  • 259. Boers SN, Van Delden JJM, Bredenoord AL. Organoids as hybrids: Ethical implications for the exchange of human tissues. J Med Ethics 2018;45(2):131–139; doi: 10.1136/medethics-2018-104846 [DOI] [PubMed] [Google Scholar]
  • 260. The Glossary for Cell & Gene Therapy and Regenerative Medicine 2020. Regen Med 2020;15(12S):1–170; doi: 10.2217/rme-2020-1512s [DOI] [PubMed] [Google Scholar]
  • 261. Mahtani KR, Heneghan C, Aronson J. Single screening or double screening for study selection in systematic reviews? BMJ Evid Based Med 2020;25(4):149–150; doi: 10.1136/bmjebm-2019-111269 [DOI] [PubMed] [Google Scholar]
  • 262. McDougall R. Systematic reviews in bioethics: Types, challenges, and value. J Med Philos (United Kingdom) 2014;39(1):89–97; doi: 10.1093/jmp/jht059 [DOI] [PubMed] [Google Scholar]
  • 263. Iglesias-López C, Agustí A, Obach M, et al. Regulatory framework for advanced therapy medicinal products in Europe and United States. Front Pharmacol 2019;10:1–14; doi: 10.3389/fphar.2019.00921 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental data
Supp_DataS1.pdf (169KB, pdf)
Supplemental data
Supp_DataS2.pdf (124.1KB, pdf)
Supplemental data
Supp_DataS3.pdf (772.8KB, pdf)

Articles from Tissue Engineering. Part B, Reviews are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES