Advertisement
REVIEW| Volume 46, ISSUE 3, P543-565, March 2023

Download started.

Ok

Ovarian rescue in women with premature ovarian insufficiency: facts and fiction

Published:December 19, 2022DOI:https://doi.org/10.1016/j.rbmo.2022.12.011

      Abstract

      The ovary has a comparatively short functional lifespan compared with other organs, and genetic and pathological injuries can further shorten its functional life. Thus, preserving ovarian function should be considered in the context of women with threats to ovarian reserve, such as ageing, premature ovarian insufficiency (POI) and diminished ovarian reserve (DOR). Indeed, one-third of women with POI retain resting follicles that can be reactivated to produce competent oocytes, as proved by the in-vitro activation of dormant follicles. This paper discusses mechanisms and clinical data relating to new therapeutic strategies using ovarian fragmentation, stem cells or platelet-rich plasma to regain ovarian function in women of older age (>38 years) or with POI or DOR. Follicle reactivation techniques show promising experimental outcomes and have been successful in some cases, when POI is established or DOR diagnosed; however, there is scarce clinical evidence to warrant their widespread clinical use. Beyond these contexts, also discussed is how new insights into the biological mechanisms governing follicular dynamics and oocyte competence may play a role in reversing ovarian damage, as no technique modifies oocyte quality. Additional studies should focus on increasing follicle number and quality. Finally, there is a small but important subgroup of women lacking residual follicles and requiring oocyte generation from stem cells.

      Key words

      Introduction

      Health is one of the fundamental principles and rights of human beings. While the last century was characterized by population growth, the 21st century is predicted to be one of population ageing. By 2050, an estimated 30% of the population will be over the age of 65 (

      Eurostat 2020. Population projections in the EU. https://ec.europa.eu

      ). With this population ageing comes an increased demand for anti-ageing approaches aided by an identification of clear hallmarks of ageing (
      • López-Otín C
      • Blasco MA
      • Partridge L
      • Serrano M
      • Kroemer G.
      The hallmarks of aging.
      ) and the knowledge that overall functional decline is slow but steady. Yet, the ovary is unique in that this organ has an abrupt reduction in function that reaches extinction within roughly 10 years, while other organs remain fully functional. The ovarian ageing phenomenon could be caused by the high demand of the physiological requirements needed for function, leading to diminished ovarian reserve (DOR) and increased oocyte/embryo aneuploidy (
      • Richardson RB
      • Allan DS
      • Le Y
      Greater organ involution in highly proliferative tissues associated with the early onset and acceleration of ageing in humans.
      ), as well as reduced ovarian volume, increased fibrosis and loss of ovarian structure (
      • Nicosia SV.
      The aging ovary.
      ), common features of physiological ageing found in young women with premature ovarian insufficiency (POI).
      POI is a heterogeneous condition affecting 1% of women of reproductive age and is defined by elevated gonadotrophins, including FSH, and irregular or absent menstruation before the age of 40 years (
      • Tucker E
      • Grover EJ
      • Bachelot A
      • Touraine P
      • Sinclair AH.
      Premature Ovarian Insufficiency: New Perspectives on Genetic Cause and Phenotypic Spectrum.
      ). DOR or poor ovarian response to stimulation is also heterogeneous and is frequently encountered during fertility treatment in regularly menstruating women (
      • Devine K
      • Mumford SL
      • Wu M
      • DeCherney AH
      • Hill MJ
      • Propst A
      Diminished ovarian reserve in the United States assisted reproductive technology population: diagnostic trends among 181,536 cycles from the Society for Assisted Reproductive Technology Clinic Outcomes Reporting System.
      ). Many – but not all – cases of DOR observed in clinical practice are physiological and represent an age-dependent decline in ovarian reserve. However, a common feature of all these clinical conditions is the reduced reproductive potential to achieve motherhood with the woman's own eggs.
      The possibility of activating dormant follicles within the ovarian cortex piqued interest after their reactivation to grow and produce healthy offspring was shown by Li and colleagues using an in-vitro activation (IVA) protocol for dormant follicles involving PTEN (phosphatase and tensin homolog) inhibitors and PI3K (phosphatidylinositol 3 kinase) stimulators (Akt [serine/threonine protein kinase 1] stimulation) (
      • Li J
      • Kawamura K
      • Cheng Y
      • Liu S
      • Klein C
      • Duan EK
      • Hsueh AJ.
      Activation of dormant ovarian follicles to generate mature eggs.
      ). This finding can be considered as the stepping stone to ovarian reactivation techniques, together with the demonstration in ovarian biopsies from women with POI that almost 40% retained some (30%) or many (9%) follicles in the cortex (
      • Van Kasteren YM
      • Schoemaker J.
      Premature ovarian failure: A systematic review on therapeutic interventions to restore ovarian function and achieve pregnancy.
      ).
      However, when considering the efficacy of the proposed reactivation techniques, it is important to highlight that large observational studies (
      • Bachelot A
      • Nicolas C
      • Bidet M
      • Dulon J
      • Leban M
      • Golmard JL
      • Polak M
      • Touraine P
      Long-term outcome of ovarian function in women with intermittent premature ovarian insufficiency.
      ;
      • Bidet M
      • Bachelot A
      • Bissauge E
      • Golmard JL
      • Gricourt S
      • Dulon J
      • Coussieu C
      • Badachi Y
      • Touraine P.
      Resumption of ovarian function and pregnancies in 358 patients with premature ovarian failure.
      ) have found that the ovary can spontaneously re-start functioning and even achieve naturally conceived pregnancies. In particular, there seems to be a huge difference in prognosis among individuals with high FSH concentrations and long periods of amenorrhoea, where naturally conceived pregnancies have been described in 4.4% of cases, and those with fluctuating POI (slightly elevated serum FSH, sporadic spotting), who have a 17.5% naturally conceived pregnancy rate (
      • Bachelot A
      • Nicolas C
      • Bidet M
      • Dulon J
      • Leban M
      • Golmard JL
      • Polak M
      • Touraine P
      Long-term outcome of ovarian function in women with intermittent premature ovarian insufficiency.
      ;
      • Bidet M
      • Bachelot A
      • Bissauge E
      • Golmard JL
      • Gricourt S
      • Dulon J
      • Coussieu C
      • Badachi Y
      • Touraine P.
      Resumption of ovarian function and pregnancies in 358 patients with premature ovarian failure.
      ). Thus, although the best clinical model to test alternatives to activate resting follicles is POI, the important number of patients with DOR and their demands to avoid the use of third-party reproduction has forced researchers and clinicians to apply the same principles of follicular activation in women with POI to those with DOR in an attempt to circumvent their damaged follicular pool. Because of the different aetiologies, POI and DOR are not comparable and perhaps experience will bring an understanding of which method of activating follicles works in which condition.
      This review discusses the techniques available, the potential indications and the outcomes of studies aimed at follicular rescue and ovarian reactivation via prolonging reproductive function. It also discusses their advantages and limitations for future use in the clinical setting and new innovative scientific developments that promise major changes in the field.

      Search methods

      PubMed was searched for peer-reviewed original English-language manuscripts published in the last 10 years to identify studies on established and experimental techniques proposed to restore ovarian reproductive function. Conditions of interest were POI, DOR, poor ovarian response (POR) and older age. The following keywords were used: oocyte ageing, fertility preservation, ovarian reserve, anti-Müllerian hormone (AMH), antral follicle count (AFC), resting/dormant follicles, follicular rescue, IVA, ovarian fragmentation, stem cells, bone marrow-derived stem cells (BMDSC), platelet-rich plasma (PRP) and autologous stem cell ovarian transplantation (ASCOT), in various combinations. The study also included relevant papers cited within these primary references. The literature was screened to identify experimental data regarding ovarian function and oocyte quality with potential translational applications to expand ovarian lifespan. Keywords such as spindle transfer, soluble growth factors, DNA damage and repair, double-strand breaks, apoptosis, telomeres, oocyte generation, stem cells and ovarian ageing were searched alone and in different combinations.
      Full articles, including clinical trials (randomized or non-randomized), case reports and case series, were retrieved and reviewed. In addition, registered clinical trials (from www.clinicaltrials.gov) were reviewed to explore indications and alternative methods for expanding the function of human ovaries after injury or at a patient's request. A total of 207 original references were included in this review.

      Pathways to induce follicular activation and ovarian rescue

      Folliculogenesis in mammals starts with the activation of a few dormant primordial follicles (around 1000 /month) to initiate growth (
      • Zhang H
      • Risal S
      • Gorre N
      • Busayavalasa K
      • Li X
      • Shen Y
      • Bosbach B
      • Brannstrom M
      • Liu K.
      Somatic cells initiate primordial follicle activation and govern the development of dormant oocytes in mice.
      ) after menarche. The transition from primordial to primary stage is characterized by both oocyte growth and the differentiation of flattened pregranulosa cells into proliferative cuboidal granulosa cells. Follicles then progress through different developmental stages to finally undergo ovulation or atresia. Although a developmental programme intrinsic to the oocyte is crucial for orchestrating follicular growth rate, whether activation of the oocyte and/or pregranulosa cells is coordinated remains unclear (
      • Adhikari D
      • Liu K.
      Molecular mechanisms underlying the activation of mammalian primordial follicles.
      ;
      • Castrillon DH
      • Miao L
      • Kollipara R
      • Horner JW
      • DePinho RA.
      Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a.
      ;
      • Eppig JJ
      • Wigglesworth K
      • Pendola FL
      The mammalian oocyte orchestrates the rate of ovarian follicular development.
      ;
      • John GB
      • Gallardo TD
      • Shirley LJ
      • Castrillon DH
      Foxo3 is a PI3K-dependent molecular switch controlling the initiation of oocyte growth.
      ;
      • Reddy P
      • Liu L
      • Adhikari D
      • Jagarlamudi K
      • Rajareddy S
      • Shen Y
      • Du C
      • Tang W
      • Hamalainen T
      • Peng SL
      • Lan ZJ
      • Cooney AJ
      • Huhtaniemi I
      • Liu K
      Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool.
      ).
      Several signalling pathways (Supplementary Table) are involved in follicle activation guidance through the control of oocyte growth initiation and maintenance, and mediating the primordial or primary transition, such as PTEN deletion on chromosome 10, PI3K, FOXO3 (the transcription factor forkhead box O3) and mTORC1 (the mammalian target of rapamycin complex 1). Indeed, knockout mouse models with an oocyte-specific deletion of Pten or Foxo3 showed a reduced reproductive lifespan due to the activation of all dormant follicles (
      • Castrillon DH
      • Miao L
      • Kollipara R
      • Horner JW
      • DePinho RA.
      Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a.
      ;
      • Reddy P
      • Liu L
      • Adhikari D
      • Jagarlamudi K
      • Rajareddy S
      • Shen Y
      • Du C
      • Tang W
      • Hamalainen T
      • Peng SL
      • Lan ZJ
      • Cooney AJ
      • Huhtaniemi I
      • Liu K
      Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool.
      ). PI3K signalling involves kinases, phosphatases and transcription factors regulating cell proliferation, metabolism, migration and survival (
      • Blume-Jensen P
      • Hunter T.
      Oncogenic kinase signalling.
      ) and is considered the primary regulator of activation. PTEN inhibition promotes the activation of PI3K to phosphorylate intermediate effectors including Akt to finally induce the nuclear export of downstream FoxO3 proteins, leading to primordial follicle activation (
      • John GB
      • Gallardo TD
      • Shirley LJ
      • Castrillon DH
      Foxo3 is a PI3K-dependent molecular switch controlling the initiation of oocyte growth.
      ).
      The interactions between the ovarian extracellular matrix and the follicles also seem to play a paramount role in follicular activation (
      • Hsueh AJW
      • Kawamura K.
      Hippo signaling disruption and ovarian follicle activation in infertile patients.
      ;
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ;
      • Shah JS
      • Sabouni R
      • Cayton Vaught KC
      • Owen CM
      • Albertini DF
      • Segars JH
      Biomechanics and mechanical signaling in the ovary: a systematic review.
      ). The granulosa cells have rigidity sensors in the form of transmembrane integrins that are coupled to intracellular proteins showing Rho-GTPase (
      • Provenzano PP
      • Keely PJ.
      Mechanical signaling through the cytoskeleton regulates cell proliferation by coordinated focal adhesion and Rho GTPase signaling.
      ) and ROCK (Rho-associated protein kinase) activity. The integrity of the extracellular matrix also results in intracellular actin de-polymerization (
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ). This maintains phosphorylation of the transcription factor YAP (Yes1 associated transcriptional regulator), preventing it from moving from the cytoplasm to the nucleus. This is known as the Salvador–Hippo–Warts (SHW) pathway (
      • Pan D.
      Hippo signaling in organ size control.
      ). When the rigidity of the extracellular matrix decreases (for example, after ovarian tissue fragmentation), actin polymerization increases in the granulosa cells and Rho and ROCK activity decreases, leading to a lower degree of YAP phosphorylation and resulting in an interruption of the intracellular SHW signalling (
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ). Migration of phosphorylated YAP to the nucleus promotes the expression of CCNs (growth promoters, namely CYR61 [cysteine-rich angiogenic protein 61 or CCN1], CTGF [connective tissue growth factor or CCN2] and NOV [nephroblastoma overexpressed or CCN3]) and BIRC (baculoviral inhibitors of apoptosis repeat containing, which are apoptosis inhibitors), and subsequently promotes small secondary follicle growth and granulosa cell formation in addition to primordial activation (
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ).

      Techniques to induce ovarian rescue in humans: basic experimental studies and clinical data in DOR, POI and older women

      IVA and ovarian fragmentation

      The findings described above prompted a proposed protocol for the IVA of dormant follicles using PTEN inhibitors and PI3K stimulators (Akt stimulation) over 48 h in mice and human ovaries (
      • Li J
      • Kawamura K
      • Cheng Y
      • Liu S
      • Klein C
      • Duan EK
      • Hsueh AJ.
      Activation of dormant ovarian follicles to generate mature eggs.
      ). Indeed, Akt signalling promoted the development of primordial follicles while also stimulating secondary follicles (
      • Fan HY
      • Liu Z
      • Cahill N
      • Richards JS.
      Targeted disruption of Pten in ovarian granulosa cells enhances ovulation and extends the life span of luteal cells.
      ;
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ). This can be considered as the proof of concept for oocyte/ovarian reactivation because it demonstrates that residual dormant follicles, remaining in the ovaries when ovarian function is lacking, can be rescued to extend the reproductive lifespan of diseased ovaries.
      Kawamura and colleagues improved the former IVA technique by including inhibition of the Hippo pathway to promote follicle activation, bringing about a synergic effect in different follicle populations, and increasing development to antral stages to produce mature oocytes and embryos to transfer (
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ) (Figure 1).
      Figure 1
      Figure 1Mechanisms of ovarian fragmentation and in-vitro activation (IVA) of dormant follicles. (A) Ovarian fragmentation for follicle activation (OFFA). (B) IVA with Akt stimulators. bpV(pic), dipotassium bisperoxo (picolinato) oxovanadate (V); OC, ovarian cortex. Created with BioRender.com.
      Increased rigidity is a common feature of menopausal ovaries, together with reduced ovarian volume, increased fibrosis and loss of ovarian structure (
      • Nicosia SV.
      The aging ovary.
      ). These characteristics can be found in both physiological ageing and in young women with POI. Since the disruption of the extracellular matrix results in growth factors being released after Hippo inhibition, it has been postulated that the disruption of the extracellular matrix can also stimulate somatic cell proliferation and therefore contribute to improving the ovarian niche to support follicle development (
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ). This theory is further supported by the fact that in animal models, the use of antifibrotic drugs for pulmonary fibrosis allowed a reversal of age-associated ovarian fibrosis while improving ovulation and extending fertility (
      • Umehara T
      • Winstanley YE
      • Andreas E
      • Morimoto A
      • Williams EJ
      • Smith KM
      • Carroll J
      • Febbraio MA
      • Shimada M
      • Russell DL
      • Robker RL
      Female reproductive life span is extended by targeted removal of fibrotic collagen from the mouse ovary.
      ).

      Clinical data from women with DOR or POI

      Since the first manuscript describing the technique for women with POI that consists of mechanical removal of the ovarian cortex via laparoscopy and subsequent fragmentation into 1 mm3 cubes to inhibit the Hippo pathway followed by IVA over 48 h with Akt stimulators before tissue autografting into the patient via laparoscopy (Figure 1), many case series and cohort studies using the same approach have reported varying degrees of success in women with DOR for POI (
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ;
      • Suzuki N
      • Yoshioka N
      • Takae S
      • Sugishita Y
      • Tamura M
      • Hashimoto S
      • Morimoto Y
      • Kawamura K.
      Successful fertility preservation following ovarian tissue vitrification in patients with primary ovarian insufficiency.
      ;
      • Zhai J
      • Yao G
      • Dong F
      • Bu Z
      • Cheng Y
      • Sato Y
      • Hu L
      • Zhang Y
      • Wang J
      • Dai S
      • Li J
      • Sun J
      • Hsueh AJ
      • Kawamura K
      • Sun Y
      In Vitro Activation of Follicles and Fresh Tissue Auto-transplantation in Primary Ovarian Insufficiency Patients.
      ). Based on the authors’ experience with fertility preservation for oncological patients, for which the surgical procedures are similar to those of IVA, except for the culture with Akt stimulators, it was learned that oncological patients in whom the ovarian cortex was fragmented into small pieces and subsequently reimplanted performed much better in terms of revascularization, return to cycling and pregnancy outcomes compared with women whose entire cortex was first removed and subsequently reimplanted as a bigger fragment (
      • Diaz-Garcia C
      • Domingo J
      • Garcia-Velasco JA
      • Herraiz S
      • Mirabet V
      • Iniesta I
      • Cobo A
      • Remohi J
      • Pellicer A.
      Oocyte vitrification versus ovarian cortex transplantation in fertility preservation for adult women undergoing gonadotoxic treatments: a prospective cohort study.
      ).
      With this background, the authors modified the IVA technique to what was referred to as ovarian fragmentation for follicular activation (OFFA), which was subsequently also employed as ‘drug-free IVA’ (
      • Ferreri J
      • Fàbregues F
      • Calafell JM
      • Solernou R
      • Borrás A
      • Saco A
      • Manau D
      • Carmona F.
      Drug-free in-vitro activation of follicles and fresh tissue autotransplantation as a therapeutic option in patients with primary ovarian insufficiency.
      ;
      • Ferreri J
      • Méndez M
      • Calafell JM
      • Fábregues F
      Long-term outcome of ovarian function after drug-free in vitro activation (IVA) in primary ovarian insufficiency patient.
      ;
      • Kawamura K
      • Ishizuka B
      • Hsueh AJW.
      Drug-free in-vitro activation of follicles for infertility treatment in poor ovarian response patients with decreased ovarian reserve.
      ;
      • Lunding SA
      • Pors SE
      • Kristensen SG
      • Landersoe SK
      • Jeppesen JV
      • Flachs EM
      • Pinborg A
      • Macklon KT
      • Pedersen AT
      • Andersen CY
      • Andersen AN.
      Biopsying, fragmentation and autotransplantation of fresh ovarian cortical tissue in infertile women with diminished ovarian reserve.
      ;
      • Méndez M
      • Fabregues F
      • Ferreri J
      • Calafell JM
      • Villarino A
      • Otero J
      • Farre R
      • Carmona F.
      Biomechanical characteristics of the ovarian cortex in POI patients and functional outcomes after drug-free IVA.
      ;
      • Tanaka Y
      • Hsueh AJ
      • Kawamura K.
      Surgical approaches of drug-free in vitro activation and laparoscopic ovarian incision to treat patients with ovarian infertility.
      ) (Figure 1A). This new approach reduces the number of operations required while avoiding ovarian tissue cryopreservation and associated follicular loss. This drug-free approach is more readily applicable to clinical practice due to the potential carcinogenic effects of PTEN inhibition (
      • Mukherjee R
      • Vanaja KG
      • Boyer JA
      • Gadal S
      • Solomon H
      • Chandarlapaty S
      • Levchenko A
      • Rosen N.
      Regulation of PTEN translation by PI3K signaling maintains pathway homeostasis.
      ), and also to the compromised further in-vitro development and DNA damage response reported in primordial oocytes after Akt stimulation (
      • McLaughlin M
      • Kinnell HL
      • Anderson RA
      • Telfer EE.
      Inhibition of phosphatase and tensin homologue (PTEN) in human ovary in vitro results in increased activation of primordial follicles but compromises development of growing follicles.
      ;
      • Maidarti M
      • Clarkson YL
      • McLaughlin M
      • Anderson RA
      • Telfer EE.
      Inhibition of PTEN activates bovine non-growing follicles in vitro but increases DNA damage and reduces DNA repair response.
      ).
      The overall cumulative pregnancy rate ranged from a success rate as high as 30% in a well-characterized population with POI to 60% in women with DOR whose inclusion criteria were solely based on repeated serum AMH concentrations of ≤5 pmol/l (0.7 ng/ml) (
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ;
      • Lunding SA
      • Pors SE
      • Kristensen SG
      • Landersoe SK
      • Jeppesen JV
      • Flachs EM
      • Pinborg A
      • Macklon KT
      • Pedersen AT
      • Andersen CY
      • Andersen AN.
      Biopsying, fragmentation and autotransplantation of fresh ovarian cortical tissue in infertile women with diminished ovarian reserve.
      ). The heterogeneity in reported results could be in part due to differences in the definition of the primary end-points between studies: while some used the number of follicles in response to stimulation (
      • Lunding SA
      • Pors SE
      • Kristensen SG
      • Landersoe SK
      • Jeppesen JV
      • Flachs EM
      • Pinborg A
      • Macklon KT
      • Pedersen AT
      • Andersen CY
      • Andersen AN.
      Biopsying, fragmentation and autotransplantation of fresh ovarian cortical tissue in infertile women with diminished ovarian reserve.
      ), others did not use predefined outcomes to assess the success of the technique. The protocols used to activate follicles also varied between experimental settings (Table 1). Other limitations of the above-described studies include the low number of participants included or the lack of a control group.
      Table 1Studies reporting the effect of ovarian fragmentation, in POR, POI, premenopausal, and postmenopausal patients.
      InterventionDiagnosis and age of patientsStudy designOutcomes overall
      Unilateral or bilateral ovarian fragmentation, ovarian tissue vitrification, warming + incubation with bpV(HOpic) + 740-YP and reimplantation of the tissue during the same surgical procedure to the serosa of both Fallopian tubes (
      • Kawamura K
      • Cheng Y
      • Suzuki N
      • Deguchi M
      • Sato Y
      • Takae S
      • Ho CH
      • Kawamura N
      • Tamura M
      • Hashimoto S
      • Sugishita Y
      • Morimoto Y
      • Hosoi Y
      • Yoshioka N
      • Ishizuka B
      • Hsueh AJ.
      Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment.
      ;
      • Suzuki N
      • Yoshioka N
      • Takae S
      • Sugishita Y
      • Tamura M
      • Hashimoto S
      • Morimoto Y
      • Kawamura K.
      Successful fertility preservation following ovarian tissue vitrification in patients with primary ovarian insufficiency.
      )
      POI <40 years oldProspective cohort study (n = 37)9/37 developed follicles

      3/37 achieved a biochemical pregnancy

      2/37 had a live birth
      Unilateral ovarian fragmentation + incubation with bpV(HOpic) + 740-YP and reimplantation of the tissue during the same surgical procedure to the serosa of both Fallopian tubes (
      • Zhai J
      • Yao G
      • Dong F
      • Bu Z
      • Cheng Y
      • Sato Y
      • Hu L
      • Zhang Y
      • Wang J
      • Dai S
      • Li J
      • Sun J
      • Hsueh AJ
      • Kawamura K
      • Sun Y
      In Vitro Activation of Follicles and Fresh Tissue Auto-transplantation in Primary Ovarian Insufficiency Patients.
      )
      POI

      29 years old (23–35)

      3.9 years of menopause (1–6)

      All undergoing IVF
      Prospective cohort study (n = 14)6/14 patients developed follicles

      4/14 had embryos frozen

      1/14 had a live birth
      Unilateral ovarian fragmentation (without incubation) and reimplantation to a peritoneal pocket (
      • Lunding SA
      • Pors SE
      • Kristensen SG
      • Landersoe SK
      • Jeppesen JV
      • Flachs EM
      • Pinborg A
      • Macklon KT
      • Pedersen AT
      • Andersen CY
      • Andersen AN.
      Biopsying, fragmentation and autotransplantation of fresh ovarian cortical tissue in infertile women with diminished ovarian reserve.
      )
      POR and infertility 37 years old (30–39)

      Preserved menstrual cycles

      AMH ≤5 pmol/l
      Prospective cohort study (n = 20)7/20 experienced elevation of AMH

      12/20 resulted in a clinical pregnancy without ART

      16/20 started at least one IVF cycle (38 cycles performed in total)

      7/20 had a live birth after IVF (18.4% cumulative LBR per IVF cycle)

      1/20 had a live birth after IUI (+1 termination)

      3/20 had a live birth after natural conception
      Unilateral ovarian fragmentation (without incubation) and reimplantation to ovary (
      • Kawamura K
      • Ishizuka B
      • Hsueh AJW.
      Drug-free in-vitro activation of follicles for infertility treatment in poor ovarian response patients with decreased ovarian reserve.
      )
      POR and infertility

      34 years (30–45)

      AMH 0–0.8 n
      g/ml

      All intended to undergo IVF
      Case series (n = 11)9/11 increased AMH levels (baseline prior to the procedure)

      3/11 resulted in a clinical pregnancy after IVF (1 miscarriage, 1 ongoing, 1 live birth)

      1/11 resulted in a natural conception (ongoing)
      Unilateral ovarian fragmentation (without incubation) and reimplantation to the contralateral ovary and peritoneal pocket (
      • Ferreri J
      • Fàbregues F
      • Calafell JM
      • Solernou R
      • Borrás A
      • Saco A
      • Manau D
      • Carmona F.
      Drug-free in-vitro activation of follicles and fresh tissue autotransplantation as a therapeutic option in patients with primary ovarian insufficiency.
      • Ferreri J
      • Méndez M
      • Calafell JM
      • Fábregues F
      Long-term outcome of ovarian function after drug-free in vitro activation (IVA) in primary ovarian insufficiency patient.
      )
      POI

      33 years (29–36 years)

      All intended to undergo IVF
      Prospective cohort study (n = 14)4/14 resulted in a clinical pregnancy after IVF
      Unilateral ovarian fragmentation (without incubation) and reimplantation to ovary (
      • Díaz-García C
      • Herraiz S
      • Pamplona M
      • Subirá J
      • Soriano MJ
      • Simon C
      • Seli E
      • Pellicer A.
      Follicular Activation in women previously diagnosed with poor ovarian response: a randomized controlled trial.
      )
      POR and infertility

      Previous failed IVF

      36 years (34–39)

      AMH 0-0.6
      g/ml

      All intended to undergo IVF
      Randomized controlled trial (n = 34)

      Control arm: no intervention
      Higher AFC after intervention in the operated ovary (difference 2 antral follicles, P = 0.021)

      Clinical pregnancy rate after IVF 2/15 with intervention versus 3/16 controls

      Live birth rate after IVF 1/15 with intervention versus 3/16 controls
      AFC, antral follicle count; AMH, anti-Müllerian hormone; ART, assisted reproductive technology; bpV(HOpic), Bisperoxovanadium(HOpic), Dipotassium bisperoxo (5-hydroxypyridine-2-carboxyl) oxovanadate (V); IUI, intrauterine insemination; LBR, live birth rate; POI, premature ovarian insufficiency; POR, poor ovarian response.
      * values for age of patients and duration of infertility are shown as mean and (range).
      Recently, the current authors’ group published the results of a randomized controlled trial in which the efficacy of OFFA to increase the number of antral follicles was assessed in women with DOR (
      • Díaz-García C
      • Herraiz S
      • Pamplona M
      • Subirá J
      • Soriano MJ
      • Simon C
      • Seli E
      • Pellicer A.
      Follicular Activation in women previously diagnosed with poor ovarian response: a randomized controlled trial.
      ), according to European Society for Human Reproduction and Embryology (ESHRE) criteria (
      • Ferraretti A.P.
      • la Marca A.
      • Fauser B.C.J.M.
      • Tarlatzis B.
      • Nargund G.
      • Gianaroli L.
      ESHRE consensus on the definition of ’poor response to ovarian stimulation for in vitro fertilization: The Bologna criteria.
      ). Although OFFA resulted in an inhibition of the Hippo pathway, confirmed by an 18.8% reduction in the phospho-YAP/YAP ratio and BIRC and CCN overexpression after fragmentation, and an increase in AFC (P = 0.021), serum AMH or FSH concentrations did not improve. Indeed, reproductive and IVF cycle outcomes were not statistically different between the groups.
      More research is needed in this field, but given evidence of a molecular and biological effect, patients for whom egg donation is not an alternative treatment and in whom follicles are very unlikely to develop (individuals with POI) may have follicular activation through fragmentation, which could still play a role in treatment. Indeed, clinicaltrials.gov reveals several ongoing studies evaluating this technique, and new modifications of the procedure, for both DOR and POI patients (NCT04608695, NCT04390308, NCT03670407, NCT03298750 and NCT02322060).

      The use of stem cells

      Up to 20 different types of adult stem cell are found in human tissues with the capacity to differentiate into the cells of the tissue in which they were originally located (
      • Lim WF
      • Inoue-Yokoo T
      • Tan KS
      • Lai MI
      • Sugiyama D.
      Hematopoietic cell differentiation from embryonic and induced pluripotent stem cells.
      ). Among them, both haemopoietic and mesenchymal cells have become increasingly important in regenerative medicine, with mesenchymal stem cells (MSC) popularly tested due to a higher replication potential and the capability to differentiate into distinct cells (
      • Friedenstein AJ
      • Petrakova KV
      • Kurolesova AI
      • Frolova GP.
      Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues.
      ).
      Different stem cell types from mesenchymal (MSC), endometrial, menstrual, adipose and haemopoietic origins (
      • Abd-Allah SH
      • Shalaby SM
      • Pasha HF
      • El-Shal AS
      • Raafat N
      • Shabrawy SM
      • Awad HA
      • Amer MG
      • Gharib MA
      • El Gendy EA
      • Raslan AA
      • El-Kelawy HM.
      l. Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits.
      ;
      • Lai D
      • Wang F
      • Chen Y
      • Wang L
      • Wang Y
      • Cheng W.
      Human amniotic fluid stem cells have a potential to recover ovarian function in mice with chemotherapy-induced sterility.
      ;
      • Lai D
      • Wang F
      • Yao X
      • Zhang Q
      • Wu X
      • Xiang C.
      Human endometrial mesenchymal stem cells restore ovarian function through improving the renewal of germline stem cells in a mouse model of premature ovarian failure.
      ;
      • Lee HJ
      • Selesniemi K
      • Niikura Y
      • Niikura T
      • Klein R
      • Dombkowski DM
      • Tilly JL
      Bone marrow transplantation generates immature oocytes and rescues long-term fertility in a preclinical mouse model of chemotherapy-induced premature ovarian failure.
      ;
      • Wang Z
      • Wang Y
      • Yang T
      • Li J
      • Yang X
      Study of the reparative effects of menstrual-derived stem cells on premature ovarian failure in mice.
      ) can restore ovarian function in chemotherapy-induced POI mouse models. Umbilical cord cells (
      • Lv X
      • Guan C
      • Li Y
      • Su X
      • Zhang L
      • Wang X
      • Xia HF
      • Ma X
      Effects of single and multiple transplantations of human umbilical cord mesenchymal stem cells on the recovery of ovarian function in the treatment of premature ovarian failure in mice.
      ;
      • Mohamed SA
      • Shalaby SM
      • Abdelaziz M
      • Brakta S
      • Hill WD
      • Ismail N
      • Al-Hendy A
      Human Mesenchymal Stem Cells Partially Reverse Infertility in Chemotherapy-Induced Ovarian Failure.
      ;
      • Shareghi-oskoue O
      • Aghebati-Maleki L
      • Yousefi M.
      Transplantation of human umbilical cord mesenchymal stem cells to treat premature ovarian failure.
      ;
      • Song D
      • Zhong Y
      • Qian C
      • Zou Q
      • Ou J
      • Shi Y
      • Gao L
      • Wang G
      • Liu Z
      • Li H
      • Ding H
      • Wu H
      • Wang F
      • Wang J
      • Li H.
      Human umbilical cord mesenchymal stem cells therapy in cyclophosphamide-induced premature ovarian failure rat model.
      ;
      • Zhang M
      • Xie T
      • Dai W
      • Zhao B
      • Zheng Y
      • Hu J
      • Pan R
      • Wang L.
      Umbilical Cord Mesenchymal Stem Cells Ameliorate Premature Ovarian Insufficiency in Rats.
      ;
      • Zheng Q
      • Fu X
      • Jiang J
      • Zhang N
      • Zou L
      • Wang W
      • Ding M
      • Chen H
      Umbilical Cord Mesenchymal Stem Cell Transplantation Prevents Chemotherapy-Induced Ovarian Failure via the NGF/TrkA Pathway in Rats.
      ;
      • Zhu SF
      • Hu HB
      • Xu HY
      • Fu XF
      • Peng DX
      • Su WY
      • He YL.
      Human umbilical cord mesenchymal stem cell transplantation restores damaged ovaries.
      ), amniotic epithelial and mesenchymal cells (
      • Ding C
      • Li H
      • Wang Y
      • Wang F
      • Wu H
      • Chen R
      • Lv J
      • Wang W
      • Huang B.
      Different therapeutic effects of cells derived from human amniotic membrane on premature ovarian aging depend on distinct cellular biological characteristics.
      ;
      • Guan-Yu X
      • I-Hsuan L
      • Chun-Chun C
      • Chia-Chun C
      • Yen-Hua L
      • Winston TKC
      • Shinn-Chih W
      Amniotic fluid stem cells prevent follicle atresia and rescue fertility of mice with premature ovarian failure induced by chemotherapy.
      ;
      • Liu T
      • Huang Y
      • Guo L
      • Cheng W
      • Zou G.
      CD44+/CD105+ human amniotic fluid mesenchymal stem cells survive and proliferate in the ovary long-term in a mouse model of chemotherapy-induced premature ovarian failure.
      ) and other sources such as adipose MSC (
      • Sun M
      • Wang S
      • Li Y
      • Yu L
      • Gu F
      • Wang C
      • Yao Y.
      Adipose-derived stem cells improved mouse ovary function after chemotherapy-induced ovary failure.
      ) and bone marrow MSC (
      • Abd-Allah SH
      • Shalaby SM
      • Pasha HF
      • El-Shal AS
      • Raafat N
      • Shabrawy SM
      • Awad HA
      • Amer MG
      • Gharib MA
      • El Gendy EA
      • Raslan AA
      • El-Kelawy HM.
      l. Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits.
      ) also demonstrate the ability to recover hormone synthesis, follicle development and fertility in rodent models, suggesting their role as follicle reactivating agents (
      • Gao M
      • Yu Z
      • Yao D
      • Qian Y
      • Wang Q
      • Jia R.
      Mesenchymal stem cells therapy: A promising method for the treatment of uterine scars and premature ovarian failure.
      ). However, not all MSC types may have an autologous source: for instance, cells from the umbilical cord or amniotic fluid will probably have an allogenic origin (
      • Odabas S
      • Elçin AE
      • Elçin YM.
      Isolation and characterization of mesenchymal stem cells.
      ). MSC will also require further cell culture, associated with poor cell quality and an accumulation of chromosomal instabilities (
      • Miura M
      • Miura Y
      • Padilla-Nash HM
      • Molinolo AA
      • Fu B
      • Patel V
      • Seo BM
      • Sonoyama W
      • Zheng JJ
      • Baker CC
      • Chen W
      • Ried T
      • Shi S.
      Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation.
      ;
      • Neves J
      • Sousa-Victor P
      • Jasper H.
      Rejuvenating Strategies for Stem Cell-Based Therapies in Aging.
      ), limiting their clinical use in humans.
      Accordingly, the study focused on BMDSC as an autologous and accessible source (
      • Hirschi KK
      • Goodell MA.
      Hematopoietic, vascular and cardiac fates of bone marrow-derived stem cells.
      ;
      • Zhang M
      • Huang B.
      The multi-differentiation potential of peripheral blood mononuclear cells.
      ). BMDSC are mononuclear cells, including MSC, haemopoietic stem cells and endothelial progenitors, with low immunogenicity that can self-renew and differentiate after tissue damage (cytokine liberation) (
      • Price CA.
      Mechanisms of fibroblast growth factor signaling in the ovarian follicle.
      ). Moreover, the proliferation and mobilization of clinically relevant cell numbers into the peripheral blood is achieved by employing pharmacological treatments such as granulocyte colony-stimulating factor (G-CSF), and BMDSC can easily be collected by apheresis, avoiding invasive techniques.
      As adult stem cells, they can secrete soluble factors, such as cytokines and growth factors, to stimulate neighbouring cells (Figure 2A) (
      • Gnecchi M
      • Zhang Z
      • Ni A
      • Dzau VJ
      Paracrine mechanisms in adult stem cell signaling and therapy.
      ). Thus, BMDSC paracrine signalling may have a key role in the ovary (
      • Gnecchi M
      • He H
      • Liang OD
      • Melo LG
      • Morello F
      • Mu H
      • Noiseux N
      • Zhang L
      • Pratt RE
      • Ingwall JS
      • et al.
      Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells.
      ;
      • Gnecchi M
      • He H
      • Noiseux N
      • Liang OD
      • Zhang L
      • Morello F
      • Mu H
      • Melo LG
      • Pratt RE
      • Ingwall JS
      • Dzau VJ.
      Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement.
      ;
      • Lai D
      • Wang F
      • Yao X
      • Zhang Q
      • Wu X
      • Xiang C.
      Human endometrial mesenchymal stem cells restore ovarian function through improving the renewal of germline stem cells in a mouse model of premature ovarian failure.
      ;
      • Takahashi M
      • Li TS
      • Suzuki R
      • Kobayashi T
      • Ito H
      • Ikeda Y
      • Matsuzaki M
      • Hamano K.
      Cytokines produced by bone marrow cells can contribute to functional improvement of the infarcted heart by protecting cardiomyocytes from ischemic injury.
      ), as their secretome is enriched with bioactive factors that support ovarian function and recovery (
      • Esfandyari S
      • Chugh RM
      • Park HS
      • Hobeika E
      • Ulin M
      • Al-Hendy A
      Mesenchymal Stem Cells as a Bio Organ for Treatment of Female Infertility.
      ) and confer antiapoptotic and antifibrotic ovarian effects (
      • Wang L
      • Ying YF
      • Ouyang YL
      • Wang JF
      • Xu J.
      VEGF and bFGF increase survival of xenografted human ovarian tissue in an experimental rabbit model.
      ). This paracrine hypothesis has been supported by other studies using different sources of stem cells and their secretomes, including MSC-derived microvesicles and exosomes, to restore ovarian function in animal models (
      • Li Z
      • Zhang M
      • Zheng J
      • Tian Y
      • Zhang H
      • Tan Y
      • Li Q
      • Zhang J
      • Huang X.
      Human Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes Improve Ovarian Function and Proliferation of Premature Ovarian Insufficiency by Regulating the Hippo Signaling Pathway.
      ;
      • Ling L
      • Feng X
      • Wei T
      • Wang Y
      • Wang Y
      • Wang Z
      • Tang D
      • Luo Y
      • Xiong Z.
      Human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation improves ovarian function in rats with premature ovarian insufficiency (POI) at least partly through a paracrine mechanism.
      ;
      • Yang Z
      • Du X
      • Wang C
      • Zhang J
      • Liu C
      • Li Y
      • Jiang H.
      Therapeutic effects of human umbilical cord mesenchymal stem cell-derived microvesicles on premature ovarian insufficiency in mice.
      ) and promote proliferation and steroidogenesis in human granulosa cells (
      • Chugh RM
      • Park HS
      • Esfandyari S
      • Elsharoud A
      • Ulin M
      • Al-Hendy A
      Mesenchymal Stem Cell-Conditioned Media Regulate Steroidogenesis and Inhibit Androgen Secretion in a PCOS Cell Model via BMP-2.
      ;
      • Park HS
      • Chugh RM
      • El Andaloussi A
      • Hobeika E
      • Esfandyari S
      • Elsharoud A
      • Ulin M
      • Garcia N
      • Bilal M
      Al-Hendy A. Human BM-MSC secretome enhances human granulosa cell proliferation and steroidogenesis and restores ovarian function in primary ovarian insufficiency mouse model.
      ).
      Figure 2
      Figure 2Mechanisms of ovarian ovarian reactivation techniques (ovarian fragmentation for follicle activation). (A) Autologous ovarian stem cell transplantation (ASCOT) of bone marrow-derived stem cells (BMDSC) and infusion of secreted soluble factors. (B) Ovarian injection of platelet-rich plasma (PRP). Created with BioRender.com.
      The current group recently reported that the infusion of human BMDSC promotes human and mouse follicular growth by increasing ovarian vascularization and stromal cell proliferation, and reduces cell death in immunosuppressed animals in which conditions of POR and POI were mimicked, allowing fertility restoration resulting in natural conception (
      • Herraiz S
      • Buigues A
      • Diaz-Garcia C
      • Romeu M
      • Martinez S
      • Gomez-Segui I
      • Simon C
      • Hsueh AJ
      • Pellicer A
      Fertility rescue and ovarian follicle growth promotion by bone marrow stem cell infusion.
      ). In both cases, injected cells arrived from the animal's tail vein to the ovarian tissue by a process known as ‘homing’, where the damaged ovarian niche attracts undifferentiated stem cells, specifically from bone marrow, to maintain homeostasis (
      • Liu J
      • Zhang H
      • Zhang Y
      • Li N
      • Wen Y
      • Cao F
      • Ai H
      • Xue X.
      Homing and Restorative Effects of Bone Marrow-derived mesenchymal stem cells on cisplatin injured ovaries in rats.
      ).
      This ovarian rescue appears to be partially mediated by a paracrine mechanism (Figure 2A), as a characterization of the BMDSC secretome revealed soluble factors involved in the cell cycle, gene expression, pathways related to RhoGTPases, and Hippo among others (
      • Buigues A
      • Marchante M
      • de Miguel-Gómez L
      • Martinez J
      • Cervelló I
      • Pellicer A
      • Herraiz S.
      Stem cell-secreted factor therapy regenerates the ovarian niche and rescues follicles.
      ). The released factors, which regulate human ovarian tissue (
      • Buigues A
      • Diaz-Gimeno P
      • Sebastian-Leon P
      • Pellegrini L
      • Pellicer N
      • Pellicer A
      • Herraiz S.
      Pathways and factors regulated by bone marrow-derived stem cells in human ovarian tissue.
      ) include those involved in the PI3K–Akt signalling and actin cytoskeleton pathways, and Kit ligand to prime primordial to primary transition (
      • Parrott JA
      • Skinner MK.
      Kit-ligand/stem cell factor induces primordial follicle development and initiates folliculogenesis.
      ) through the PI3K–Akt pathway (
      • Adhikari D
      • Liu K.
      Molecular mechanisms underlying the activation of mammalian primordial follicles.
      ), and key regulators of the neoangiogenic process and vessel stability. Because follicular growth also depends on cyclic microvascular remodelling throughout the menstrual cycle (
      • Fraser HM.
      Regulation of the ovarian follicular vasculature.
      ), BMDSC could have a crucial angiogenic role in the ovary. Indeed, poor ovarian response is associated with reduced ovarian blood flow (
      • Pan HA
      • Wu MH
      • Cheng YC
      • Wu LH
      • Chang FM
      Quantification of ovarian stromal Doppler signals in poor responders undergoing in vitro fertilization with three-dimensional power Doppler ultrasonography.
      ).
      Clinically, follicular reactivation and niche regeneration could also explain several reports of fertility recovery and natural conception in infertile women after cancer treatment and bone marrow transplant (
      • Hershlag A
      • Schuster MW.
      Return of fertility after autologous stem cell transplantation.
      ;
      • Salooja N
      • Chatterjee R
      • McMillan AK
      • Kelsey SM
      • Newland AC
      • Milligan DW
      • Franklin IM
      • Hutchinson RM
      • Linch DC
      • Goldstone AH.
      Successful pregnancies in women following single autotransplant for acute myeloid leukemia with a chemotherapy ablation protocol.
      ;
      • Salooja N
      • Szydlo RM
      • Socie G
      • Rio B
      • Chatterjee R
      • Ljungman P
      • Van Lint MT
      • Powles R
      • Jackson G
      • Hinterberger-Fischer M
      • et al.
      Pregnancy outcomes after peripheral blood or bone marrow transplantation: a retrospective survey.
      ;
      • Sanders JE
      • Hawley J
      • Levy W
      • Gooley T
      • Buckner CD
      • Deeg HJ
      • Doney K
      • Storb R
      • Sullivan K
      • Witherspoon R
      • Appelbaum FR.
      Pregnancies following high-dose cyclophosphamide with or without high-dose busulfan or total-body irradiation and bone marrow transplantation.
      ;
      • Veitia RA
      • Gluckman E
      • Fellous M
      • Soulier J.
      Recovery of female fertility after chemotherapy, irradiation, and bone marrow allograft: further evidence against massive oocyte regeneration by bone marrow-derived germline stem cells.
      ). A healthy live birth was reported in a similar case (
      • Veitia RA
      • Gluckman E
      • Fellous M
      • Soulier J.
      Recovery of female fertility after chemotherapy, irradiation, and bone marrow allograft: further evidence against massive oocyte regeneration by bone marrow-derived germline stem cells.
      ); interestingly, analysis of the baby's, the mother's and the bone marrow donor's DNA revealed a genetic compatibility between the mother and the child. This finding dismissed a genetic relationship between the donor and the baby, so the probability that stem cells replenish oocytes directly is unlikely; instead, stem cells may promote regeneration of the ovarian niche, allowing the growth and rescue of existing residual follicles, as supported by several experimental studies (
      • Eggan K
      • Jurga S
      • Gosden R
      • Min IM
      • Wagers AJ
      Ovulated oocytes in adult mice derive from non-circulating germ cells.
      ;
      • Herraiz S
      • Buigues A
      • Diaz-Garcia C
      • Romeu M
      • Martinez S
      • Gomez-Segui I
      • Simon C
      • Hsueh AJ
      • Pellicer A
      Fertility rescue and ovarian follicle growth promotion by bone marrow stem cell infusion.
      ;
      • Zhang H
      • Panula S
      • Petropoulos S
      • Edsgärd D
      • Busayavalasa K
      • Liu L
      • Li X
      • Risal S
      • Shen Y
      • Shao J
      • Liu M
      • Li S
      • Zhang D
      • Zhang X
      • Gerner RR
      • Sheikhi M
      • Damdimopoulou P
      • Sandberg R
      • Douagi I
      • Gustafsson JÅ
      • Liu L
      • Lanner F
      • Hovatta O
      • Liu K.
      Adult human and mouse ovaries lack DDX4-expressing functional oogonial stem cells.
      ).

      Clinical data from stem cell-based therapies

      Within the clinical set-up of MSC, umbilical cord-derived MSC (UCMSC) have been shown to increase the ovarian function in 61 young patients (mean age 31 years, range 39-34 years) with POI, after transvaginal intra-ovarian allogenic stem cell infusion of UCMS isolated from donor healthy full-term human placental samples (
      • Yan L
      • Wu Y
      • Li L
      • Wu J
      • Zhao F
      • Gao Z
      • Liu W
      • Li T
      • Fan Y
      • Hao J
      • Liu J
      • Wang H.
      Clinical analysis of human umbilical cord mesenchymal stem cell allotransplantation in patients with premature ovarian insufficiency.
      ). This methodology resulted in an increased AFC, initiation of ovarian stimulation and overall pregnancy rate of 6.6% (4/61; three IVF and one natural conception) with no safety issues reported even when using an allogenic cell source. Positive results were increased in women with a shorter duration of amenorrhoea. Authors reported mature follicles only in ovaries receiving UCMSC. However, even within the same clinical trial, there were differences in the procedure, as some women received one transplant while others received two or three.
      Most clinical investigations of MSC have published the use of bone marrow-derived mesenchymal cells (BM-MSC) in humans after iliac crest puncture and posterior in-vitro culture (Table 2). However, the objective is to achieve the same results while adopting less invasive methods. For instance, BM-MSC were employed in 30 women with POI who were aged 40 years or less, with one group receiving a direct laparoscopic infusion into the ovarian stroma, and the other group undergoing unilateral catheterization of the ovarian artery (
      • Gabr H
      • Elkheir WA
      • El-Gazzar A.
      Autologous stem cell transplantation in patients with idiopathic premature ovarian failure.
      ). Overall, hormone improvement was observed in 86.7% of participants from both groups, while 60% showed ovulation at some point during follow-up. Only 10% of women underwent IVF cycles, and one natural conception was achieved. Comparable results were obtained in 10 women (26–33 years old) with POI after BM-MSC injection into both ovaries via laparoscopy (
      • Edessy M
      • Hosni H
      • Shady Y
      • Waf Y
      • Bakr S
      • Kamel M.
      Autologous stem cells therapy, The first baby of idiopathic premature ovarian failure.
      ).
      Table 2Studies reporting the effect of bone marrow stem cells for follicular activation in POR, POI, premenopausal and postmenopausal patients
      TechniqueDiagnosis and age of patient(s)Study designOutcomes overall
      BM-MSC: bilateral laparoscopy injection (
      • Edessy M
      • Hosni H
      • Shady Y
      • Waf Y
      • Bakr S
      • Kamel M.
      Autologous stem cells therapy, The first baby of idiopathic premature ovarian failure.
      )
      POI

      <40 years old (26–33 years)
      Pilot study (n = 10)Menstruation recovery in 2 patients

      1 live birth (pregnancy rate 10%)
      BM-MSC, 2 groups:

      1. Unilateral laparoscopic injection

      2. Intra-arterial cell infusion

      (
      • Gabr H
      • Elkheir WA
      • El-Gazzar A.
      Autologous stem cell transplantation in patients with idiopathic premature ovarian failure.
      )
      POI

      18–40 years old
      Pilot study (n = 30)26 patients showed improved hormone levels 1 month after treatment

      Ovulation in 18 women

      3 patients started IVF treatment

      1 natural conception pregnancy
      BM-MSC: bilateral laparoscopy injection (
      • Gupta S
      • Lodha P
      • Karthick MS
      • Tandulwadkar SR.
      Role of Autologous Bone Marrow-Derived Stem Cell Therapy for Follicular Recruitment in Premature Ovarian Insufficiency: Review of Literature and a Case Report of World's First Baby with Ovarian Autologous Stem Cell Therapy in a Perimenopausal Woman of Age.
      )
      Postmenopausal 45 years oldCase report (n = 1)8 weeks after the intervention, increase of AFC (from 1 to 2 follicles) and AMH (from 0.4 to 0.9 n
      g/ml)

      IVF started 8 weeks after treatment

      3 oocytes obtained, with 1 embryo

      1 live birth after frozen embryo transfer 11 months after treatment
      BM-MSC: bilateral laparoscopy injection (
      • Igboeli P
      • el Andaloussi A
      • Sheikh U
      • Takala H
      • Elsharoud A
      • McHugh A
      • Gavrilova-Jordan L
      • Levy S
      • Al-Hendy A
      Intraovarian injection of autologous human mesenchymal stem cells increases estrogen production and reduces menopausal symptoms in women with premature ovarian failure: Two case reports and a review of the literature.
      )
      Idiopathic POI

      36 and 42 years old
      Clinical trial (n = 2)Increase of oestrogen levels 6–9 months after treatment (first case from <10.9 to 26 pg/ml; second case some controls missing)

      Progressive decrease in FSH levels over the 6–9-month follow-up

      Recovery of menstruation 7 months after treatment
      BMDSC: unilateral intra-arterial cell infusion (
      • Herraiz S
      • Romeu M
      • Buigues A
      • Martinez S
      • Diaz-Garcia C
      • Gomez-Segui I
      • Martinez J
      • Pellicer N
      • Pellicer A
      Autologous stem cell ovarian transplantation to increase reproductive potential in patients who are poor responders.
      )
      POR

      ≤40 years old
      Prospective observational pilot study

      (n = 20; ongoing at time of publication: report on 17 patients)
      Improvement of ovarian reserve biomarkers in 13 women 2 weeks after treatment (AFC and AMH)

      28 IVF cycles initiated (78 days after ASCOT)

      Oocyte retrieval in 24/28 of the cycles, and embryos in 19/28 of the cycles

      5 pregnancies, 2 miscarriages and 3 live births
      BMDSC. 2 groups:

      1. Cell mobilization to peripheral blood

      2. Unilateral intra-arterial cell infusion

      (
      • Pellicer N
      • Herraiz S
      • Romeu M
      • Martinez S
      • Buiges A
      • Gomez-Seguí I
      • Martínez J
      • Pellicer A.
      Bone marrow derived stem cells restore ovarian function and fertility in women with POI: Interim report of a randomized trial comparing mobilization versus ovarian injection.
      )
      POI (still ongoing): report on 10 patients

      <38 years old
      Randomized prospective pilot study

      (n = 20)
      Follicular growth 90–140 days after treatment

      Positive response to BMDSC (AMH + FC): 66.6% in the ASCOT group, 50% in the mobilization group

      6 women started IVF, with 16 cycles initiated

      4 MII oocytes and 2 embryos obtained

      1 ongoing pregnancy, with 1 live birth
      AFC, antral follicle count; AMH, anti-Müllerian hormone, ASCOT, autologous stem cell ovarian transplantation; BMDSC, bone marrow mesenchymal stem cells; BM-MSC, bone marrow-derived mesenchymal cells; FC, follicle count; MII, metaphase II; POI, premature ovarian insufficiency; POR, poor ovarian response.
      In search of adopting less invasive methods, the current group described a BMDSC-based technique in women with DOR (
      • Herraiz S
      • Romeu M
      • Buigues A
      • Martinez S
      • Diaz-Garcia C
      • Gomez-Segui I
      • Martinez J
      • Pellicer N
      • Pellicer A
      Autologous stem cell ovarian transplantation to increase reproductive potential in patients who are poor responders.
      ). This innovative treatment received the name of ‘autologous stem cell ovarian transplantation’ (ASCOT). A total of 17 women with DOR according to the ESHRE criteria (
      • Ferraretti A.P.
      • la Marca A.
      • Fauser B.C.J.M.
      • Tarlatzis B.
      • Nargund G.
      • Gianaroli L.
      ESHRE consensus on the definition of ’poor response to ovarian stimulation for in vitro fertilization: The Bologna criteria.
      ) (≤40 years, 3–5 years of infertility) were included in the study. Initially, BMDSC were mobilized and collected from peripheral blood by apheresis after a 5-day pharmacological treatment (G-CSF). The cells were then infused by intra-arterial catheterization into one ovarian artery, leaving the other ovary as a control. During the 6-month follow-up, ovarian reserve biomarkers (AFC and AMH) were enhanced in 81.3% of the participants, but importantly, there was a significant positive association with the presence of the BMDSC-secreted growth factors fibroblast growth factor 2 (FGF-2), involved in follicle growth (
      • Price CA.
      Mechanisms of fibroblast growth factor signaling in the ovarian follicle.
      ), and thrombospondin-1 (key regulator of neoangiogenesis, associated with AMH increase) present in the plasma from apheresis.
      These women had a total of 24 previous failed IVF cycles, in which no pregnancies were achieved. After ASCOT, 88.2% of women started IVF cycles at some point during the follow-up, retrieving a total of 51 metaphase II (MII) oocytes and obtaining 36 embryos after intracytoplasmic sperm injection (ICSI). In total, the pregnancy rate after ASCOT was 33.3%, with three naturally conceived pregnancies. However, the euploidy rate was only 16%, due to the advanced maternal age of the participants.
      Different conclusions were drawn after this pilot study. First, the high pregnancy rate suggests that invasive cell collection followed by cell culture is not mandatory to obtain good reproductive results. Second, AFC increased in both the treated and the control ovary, suggesting that circulating growth factors and BMDSC have beneficial effects on the non-infused ovary, and bringing into question the requirement for direct arterial cell infusion. Furthermore, ovarian reserve biomarker improvement was observed 4 weeks after ASCOT treatment, supporting the theory that secondary and pre-antral follicles, already present at the time of the treatment, benefit from the stem cells’ properties and growth factors to regenerate the ovarian niche and stimulate the growth of the granulosa while preventing atresia. Although some antral and pre-antral follicles may be present in women with DOR, there is have a damaged ovarian niche that makes it impossible to maintain physiological requirements and follicular growth (
      • Gougeon A
      • Chainy GB.
      Morphometric studies of small follicles in ovaries of women at different ages.
      ; Richardson et al., 2014). Finally, BMDSC may increase ovarian reserve but have no effect on oocyte quality, which is directly related to age and exponentially decreases from 38 years onwards.
      A second prospective pilot study of ASCOT involving 20 women with POI (NCT03535480) according to the ESHRE POI criteria was carried out (
      • Pellicer N
      • Herraiz S
      • Romeu M
      • Martinez S
      • Buiges A
      • Gomez-Seguí I
      • Martínez J
      • Pellicer A.
      Bone marrow derived stem cells restore ovarian function and fertility in women with POI: Interim report of a randomized trial comparing mobilization versus ovarian injection.
      ). Participants were randomized into two study arms: the first group received 5 days of G-CSF treatment, with cells remaining in circulation in the peripheral blood to test whether cells and growth factors would arrive at the wounded ovaries; the second group were treated identically to the first ASCOT study (with blood apheresis, and cell infusion into one ovarian artery). Preliminary interim analysis in 10 participants revealed follicular development in both arms when compared with basal concentrations; these follicular growth waves were detected 90–140 days after treatments. Six women started IVF cycles after treatment in both arms (60%), 4 MII oocytes were retrieved and two vitrified embryos were successfully transferred, one in each study arm. The pregnancy rate was 10%, with one healthy live birth at 38 weeks of pregnancy. Half of the participants reported an improvement of vasomotor symptoms at some point during follow-up, although a significant decrease of FSH concentrations was not recorded.
      Stem cell studies in patients with POI addressed amelioration of ovarian reserve after more than 3 months of follow-up (
      • Gupta S
      • Lodha P
      • Karthick MS
      • Tandulwadkar SR.
      Role of Autologous Bone Marrow-Derived Stem Cell Therapy for Follicular Recruitment in Premature Ovarian Insufficiency: Review of Literature and a Case Report of World's First Baby with Ovarian Autologous Stem Cell Therapy in a Perimenopausal Woman of Age.
      ;
      • Igboeli P
      • el Andaloussi A
      • Sheikh U
      • Takala H
      • Elsharoud A
      • McHugh A
      • Gavrilova-Jordan L
      • Levy S
      • Al-Hendy A
      Intraovarian injection of autologous human mesenchymal stem cells increases estrogen production and reduces menopausal symptoms in women with premature ovarian failure: Two case reports and a review of the literature.
      ;
      • Pellicer N
      • Herraiz S
      • Romeu M
      • Martinez S
      • Buiges A
      • Gomez-Seguí I
      • Martínez J
      • Pellicer A.
      Bone marrow derived stem cells restore ovarian function and fertility in women with POI: Interim report of a randomized trial comparing mobilization versus ovarian injection.
      ). These patients have no stimulable antral follicles, so researchers have turned their attention to the dormant primordial follicles. They postulate that stem cells could restore the ovarian niche, allowing the residual follicle pool to successfully grow and develop (
      • García D
      • Brazal S
      • Rodríguez A
      • Prat A
      • Vassena R
      Knowledge of age-related fertility decline in women: A systematic review.
      ). This could explain why ovarian reserve biomarkers are successfully improved from 3 to 5 months after the intervention, as human primordial follicles take up to 5 months to reach the antral stage (
      • Baerwald AR
      • Adams GP
      • Pierson RA.
      Ovarian antral folliculogenesis during the human menstrual cycle: A review.
      ).
      Likewise, the objective should be the identification of less invasive treatments in women with DOR and POI. Indeed, at the time of this publication, several studies are using different stem cell sources being developed for these women (NCT04009473, NCT03877471, NCT03069209, NCT02043743, NCT01853501, NCT01742533 and NCT02603744).

      Platelet-rich plasma

      Paracrine actions and regenerative cell-independent mechanisms in ovarian tissue activate pre-existing quiescent follicles through a variety of soluble growth factors involved in follicular growth, angiogenesis, viability and ovarian response to ovarian stimulation (
      • Gnecchi M
      • Zhang Z
      • Ni A
      • Dzau VJ
      Paracrine mechanisms in adult stem cell signaling and therapy.
      ). In fact, a paracrine signalling mechanism is also the basis of the use of PRP, defined as the plasma fraction with the highest platelet concentration, at least 1 million/μl obtained by autologous blood centrifugation (
      • Lacci KM
      • Dardik A.
      Platelet-Rich Plasma: Support for Its Use in Wound Healing.
      ).
      PRP is enriched in terms of both platelets and growth factors, including transforming growth factor-β, vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), insulin-like growth factor-1, FGF-ß (
      • Mehta S
      • Watson JT.
      Platelet rich concentrate: basic science and current clinical applications.
      ) and, in smaller amounts, other bioactive compounds released from dense granules (
      • El-Sharkawy H
      • Kantarci A
      • Deady J
      • Hasturk H
      • Liu H
      • Alshahat M
      • van Dyke TE.
      Platelet-Rich Plasma: Growth Factors and Pro- and Anti-Inflammatory Properties.
      ). Platelets mediate haemostasis following trauma or local ischaemia by mechanisms such as adhesion, aggregation, thrombin generation and fibrin formation (
      • Anitua E.
      • Andia I.
      • Ardanza B.
      • Nurden P.
      • Nurden A.
      Autologous platelets as a source of proteins for healing and tissue regeneration.
      ). In addition, they play an important role in modulating cellular migration, extracellular matrix remodulation, cell proliferation, angiogenesis, cell differentiation and apoptosis (
      • Gurtner GC
      • Werner S
      • Barrandon Y
      • Longaker MT.
      Wound repair and regeneration.
      ). Platelet alpha-granules discharge a range of molecules, such as hepatocyte growth factor, stromal cell derived factor-1, adenosine diphosphate (ADP), serotonin and sphingosine-1-phosphate, among others (
      • Sills ES
      • Wood SH.
      Autologous activated platelet-rich plasma injection into adult human ovary tissue: Molecular mechanism, analysis, and discussion of reproductive response.
      ). These substances promote tissue repair, incite neoangiogenesis, have pro-inflammatory effects (Figure 2B) and activate the clearance of dead cells and the eradication of detritus (
      • Sills ES
      • Wood SH.
      Autologous activated platelet-rich plasma injection into adult human ovary tissue: Molecular mechanism, analysis, and discussion of reproductive response.
      ). PRP causes monocyte-mediated proinflammatory cytokine release (
      • El-Sharkawy H
      • Kantarci A
      • Deady J
      • Hasturk H
      • Liu H
      • Alshahat M
      • van Dyke TE.
      Platelet-Rich Plasma: Growth Factors and Pro- and Anti-Inflammatory Properties.
      ) and has been described as a ‘growth factor agonist’ (
      • Petrova N
      • Edmonds M.
      Emerging drugs for diabetic foot ulcers.
      ). PRP is applied in multiple fields, including periodontal and oral surgery, orthopaedics, aesthetics, burns treatment, ophthalmology and spinal, cardiac, and trauma surgery (
      • Collins T
      • Alexander D
      • Barkatali B.
      Platelet-rich plasma: a narrative review.
      ;
      • Lacci KM
      • Dardik A.
      Platelet-Rich Plasma: Support for Its Use in Wound Healing.
      ;).
      In the context of ovarian tissue, PRP could play an important role in folliculogenesis and ovulation by providing an appropriate environment to support follicle selection and growth (
      • Field SL
      • Dasgupta T
      • Cummings M
      • Orsi NM.
      Cytokines in ovarian folliculogenesis, oocyte maturation and luteinisation.
      ) in the same way that platelets are involved in wound repair and tissue regeneration after cell deterioration. Indeed, cyclic injury in the ovary following ovulation could be repeatedly healed by platelets (
      • Eppley B
      • Woodell JE
      • Higgins J.
      Platelet quantification and growth factor analysis from platelet-rich plasma: implications for wound healing.
      ;
      • Lacci KM
      • Dardik A.
      Platelet-Rich Plasma: Support for Its Use in Wound Healing.
      ). PRP cytokines have proangiogenic properties; for example, VEGF and PDGF are involved in the increased vascularization observed during follicular maturation until final ovulation (
      • Pauli SA
      • Tang H
      • Wang J
      • Bohlen P
      • Posser R
      • Hartman T
      • Sauer M.v.
      • Kitajewski J
      • Zimmermann RC.
      The vascular endothelial growth factor (VEGF)/VEGF receptor 2 pathway is critical for blood vessel survival in corpora lutea of pregnancy in the rodent.
      ). PRP has a proangiogenic effect on animals, illustrated by infusing PRP into ovaries with induced ischaemia (
      • Bakacak M
      • Mehmet SB
      • İnanc F
      • Yaylali A
      • Serin S
      • Attar R
      • Yildirim G
      • Yildirim O.
      Protective Effect of Platelet Rich Plasma on Experimental Ischemia/Reperfusion Injury in Rat Ovary.
      ), which show a significant increase in peritoneal VEGF. In contrast, antagonism of VEGF and PDGF in animals induces reduced vascularization and increased apoptosis, leading to impaired folliculogenesis and atresia (
      • Feng Y
      • Cui P
      • Lu X
      • Hsueh B
      • Möller Billig F
      • Zarnescu Yanez L
      • Tomer R
      • Boerboom D
      • Carmeliet P
      • Deisseroth K
      • et al.
      CLARITY reveals dynamics of ovarian follicular architecture and vasculature in three-dimensions.
      ;
      • Pascuali N
      • Scotti L
      • Abramovich D
      • Irusta G
      • di Pietro M
      • Bas D
      • Tesone M
      • Parborell F.
      Inhibition of platelet-derived growth factor (PDGF) receptor affects follicular development and ovarian proliferation, apoptosis and angiogenesis in prepubertal eCG-treated rats.
      ). PRP also plays an important role in mitogenesis, chemotaxis and extracellular matrix formation in the ovary (
      • Sills ES
      • Wood SH.
      Autologous activated platelet-rich plasma injection into adult human ovary tissue: Molecular mechanism, analysis, and discussion of reproductive response.
      ). Taken together, these findings illustrate how in-vitro and in-vivo PRP-released cytokines and proangiogenic factors (
      • Kakudo N
      • Morimoto N
      • Kushida S
      • Ogawa T
      • Kusumoto K.
      Platelet-rich plasma releasate promotes angiogenesis in vitro and in vivo.
      ) may impact folliculogenesis, especially in individuals with DOR and impaired ovarian mechanisms (
      • Atkinson L
      • Martin F
      • Sturmey RG.
      Intraovarian injection of platelet-rich plasma in assisted reproduction: Too much too soon?.
      ).
      An advantage of PRP is that it can be easily obtained by minimal intervention. Autologous peripheral blood is isolated from the patient, and centrifuged to separate PRP from platelet-poor plasma, ensuring a platelet concentration up to 10 times greater than the circulating concentration (
      • Fabi S
      • Sundaram H.
      The potential of topical and injectable growth factors and cytokines for skin rejuvenation.
      ). Various commercial PRP kits are available, providing predetermined platelet concentrations. However, the use of different PRP concentrations can lead to protocol deviations and heterogeneity in results.
      ‘Platelet activation’ is proposed as an important step to promote platelet growth factor release (
      • Sills ES
      • Wood SH.
      Autologous activated platelet-rich plasma injection into adult human ovary tissue: Molecular mechanism, analysis, and discussion of reproductive response.
      ). Although the role of platelet activation has not been fully established, platelet activation yields significant platelet degranulation with growth factor discharge and pro-inflammatory cytokine release (
      • Fabi S
      • Sundaram H.
      The potential of topical and injectable growth factors and cytokines for skin rejuvenation.
      ). Several techniques exist to activate platelets, including ADP, thrombin, collagen, calcium chloride, calcium gluconate or a combination of these substances (
      • Sills ES
      • Wood SH.
      Autologous activated platelet-rich plasma injection into adult human ovary tissue: Molecular mechanism, analysis, and discussion of reproductive response.
      ). In clinical practice, fine ultrasound-guided needles help to introduce PRP into the human ovary. PRP is ideally injected directly into the ovarian cortex, to favour penetration due to its large molecular weight (
      • Fabi S
      • Sundaram H.
      The potential of topical and injectable growth factors and cytokines for skin rejuvenation.
      ), allowing an interaction with receptors and ligands, activating inter- and intracellular signalling pathways, and consequently enhancing cell and follicular differentiation (
      • Menon SN
      • Flegg JA
      • Mccue SW
      • Schugart RC
      • Dawson RA
      • Sean McElwain DL
      Modelling the interaction of keratinocytes and fibroblasts during normal and abnormal wound healing processes.
      ).

      Clinical data on intra-ovarian PRP injection

      Although mechanistic experimental studies are limited, preclinical trials have been accomplished. Hosseini and colleagues evaluated the effect of PRP on primordial follicles, isolated from previously healthy deceased woman and embedded in a PRP-enriched 3D matrix (previously activated with thrombin), detecting follicle growth after 10 days in culture (
      • Hosseini L
      • Shirazi A
      • Naderi MM
      • Shams-Esfandabadi N
      • Borjian Boroujeni S
      • Sarvari A
      • Sadeghnia S
      • Behzadi B
      • Akhondi MM
      Platelet-rich plasma promotes the development of isolated human primordial and primary follicles to the preantral stage.
      ). Following these results, this approach has over recent years been tested in DOR, POI and post- or perimenopausal patients (
      • Herlihy NS
      • Seli E.
      The use of intraovarian injection of autologous platelet rich plasma (PRP) in patients with poor ovarian response and premature ovarian insufficiency.
      ). For a better understanding of the mechanism of action of PRP in ovarian repair and to clearly distinguish its benefits in different pathologies, the latest studies with separation of the pathologies are discussed here. However, not all investigations operate with the same PRP protocol, as different volumes are applied and different activation techniques are used, and some investigations do not use PRP activation. This could explain the heterogeneity of results even when the studied populations have similar characteristics.
      The first clinical data on PRP were reported as a pilot study in four women with more than 1 year of infertility, with at least one failed or cancelled assisted reproductive technology (ART) treatment, or amenorrhoea lasting for at least 3 months (
      • Sills ES
      • Rickers NS
      • Li X
      • Palermo GD.
      First data on in vitro fertilization and blastocyst formation after intraovarian injection of calcium gluconate-activated autologous platelet rich plasma.
      ). A significant reduction in FSH was observed with MII oocytes retrieved from all participants, which finally lead to one blastocyst per patient. One woman achieved pregnancy (still ongoing at the time of publication) and the other three women decided to freeze their embryos for future frozen embryo transfers.
      Another investigation reported a 15.8% pregnancy rate (three clinical pregnancies including two natural conceptions) after bilateral PRP infusion in 19 patients with DOR (
      • Farimani M
      • Heshmati S
      • Poorolajal J
      • Bahmanzadeh M.
      A report on three live births in women with poor ovarian response following intra-ovarian injection of platelet-rich plasma (PRP).
      ). The results showed a significant increase in the number of oocytes retrieved in ART treatments when compared with cycles before the intervention, with natural conceptions occurring 2–4 months after the PRP intervention. Similar results were obtained in a retrospective study with the same PRP protocol in 96 women with POR according to the Patient-Oriented Strategies Encompassing IndividualizeD Oocyte Number (POSEIDON) criteria for POR (
      • Alviggi C
      • Andersen CY
      • Buehler K
      • Conforti A
      • de Placido G
      • Esteves SC
      • Fischer R
      • Galliano D
      • Polyzos NP
      • Sunkara SK
      • Ubaldi FM
      • Humaidan P.
      A new more detailed stratification of low responders to ovarian stimulation: from a poor ovarian response to a low prognosis concept.
      ;
      • Farimani M
      • Nazari A
      • Mohammadi S
      • Anvari Aliabad R
      Evaluation of intra-ovarian platelet-rich plasma administration on oocytes-dependent variables in patients with poor ovarian response: A retrospective study according to the POSEIDON criteria.
      ). Although there were no significant differences in hormone concentrations after treatment, there was a significant increase in the number of oocytes retrieved and an increase in MII oocytes in all POSEIDON groups. Although the overall pregnancy rate was 14.6%, young participants within group 1 benefited most from the intervention (71.4% pregnancy rate) (
      • Alviggi C
      • Andersen CY
      • Buehler K
      • Conforti A
      • de Placido G
      • Esteves SC
      • Fischer R
      • Galliano D
      • Polyzos NP
      • Sunkara SK
      • Ubaldi FM
      • Humaidan P.
      A new more detailed stratification of low responders to ovarian stimulation: from a poor ovarian response to a low prognosis concept.
      ;
      • Farimani M
      • Nazari A
      • Mohammadi S
      • Anvari Aliabad R
      Evaluation of intra-ovarian platelet-rich plasma administration on oocytes-dependent variables in patients with poor ovarian response: A retrospective study according to the POSEIDON criteria.
      ).
      A large non-randomized clinical trial of 83 women with low ovarian reserve allocated patients into two different groups, with 46 receiving activated PRP injections for three consecutive cycles, and the rest receiving no intervention. After a 3-month follow-up, participants receiving PRP showed a significant decrease in FSH and a significant increase in AMH and AFC (compared with the control group). The biochemical and clinical pregnancy rate in the PRP group was statistically significantly higher than in the control group (26.1% versus 5.4%, P < 0.02; and 23.9% versus 5.4%, P < 0.03, respectively), as was the number of live births (5 versus 1) with no statistical differences for miscarriage rate. The researchers concluded that PRP improved the number and quality of the oocytes retrieved, which will lead to a superior quality embryo cohort (
      • Melo P
      • Navarro C
      • Jones C
      • Coward K
      • Coleman L.
      The use of autologous platelet-rich plasma (PRP) versus no intervention in women with low ovarian reserve undergoing fertility treatment: a non-randomized interventional study.
      ).
      Different retrospective studies were carried out in women with POR (
      • Barad DH
      • Albertini DF
      • Molinari E
      • Gleicher N.
      Preliminary report of intraovarian injections of autologous platelet-rich plasma (PRP) in extremely poor prognosis patients with only oocyte donation as alternative: a prospective cohort study.
      ;
      • Pacu I
      • Zygouropoulos N
      • Dimitriu M
      • Rosu G
      • Ionescu CA.
      Use of platelet-rich plasma in the treatment of infertility in poor responders in assisted human reproduction procedures.
      ;
      • Tülek F
      • Kahraman A.
      The effects of intra-ovarian autologous platelet rich plasma injection on IVF outcomes of poor responder women and women with premature ovarian insufficiency.
      ), showing promising results in the improvement of ovarian reserve biomarkers, especially in the first months post-PRP infusion (
      • Pacu I
      • Zygouropoulos N
      • Dimitriu M
      • Rosu G
      • Ionescu CA.
      Use of platelet-rich plasma in the treatment of infertility in poor responders in assisted human reproduction procedures.
      ),; there was also an increased number of oocytes and embryos (Tülek et al., 2022), and higher pregnancy rates (
      • Barad DH
      • Albertini DF
      • Molinari E
      • Gleicher N.
      Preliminary report of intraovarian injections of autologous platelet-rich plasma (PRP) in extremely poor prognosis patients with only oocyte donation as alternative: a prospective cohort study.
      ;
      • Pacu I
      • Zygouropoulos N
      • Dimitriu M
      • Rosu G
      • Ionescu CA.
      Use of platelet-rich plasma in the treatment of infertility in poor responders in assisted human reproduction procedures.
      ).
      The largest prospective clinical trial to date includes 510 women with POR (mean age 40.3, range 30-45 years) according to the POSEIDON criteria (
      • Cakiroglu Y
      • Yuceturk A
      • Karaosmanoglu O
      • Kopuk SY
      • Korun ZEU
      • Herlihy N
      • Scott RT
      • Tiras B
      • Seli E.
      Ovarian reserve parameters and IVF outcomes in 510 women with poor ovarian response (POR) treated with intraovarian injection of autologous platelet rich plasma (PRP).
      ). Ovarian reserve parameters showed a statistically significant increase in AFC, a rise in serum AMH and a decrease in serum FSH values after PRP; the improvement of ovarian reserve markers was positively correlated with the likelihood of producing at least one viable embryo. The natural conception rate was 4.3% (22/510), while the pregnancy rate after IVF was 26.6% (83/312) among women with embryo formation. Overall, PRP injection resulted in a 20.6% pregnancy rate in this cohort (105/510), with a total of 66 live births, 12 from natural conceptions and 54 after IVF. Interestingly, this study established that women aged 38 years or less benefited the most from PRP, as 40 years of age (sensitivity 48.3, specificity 70.9; area under the curve [AUC] 0.612) was the cut-off for the those who would not benefit due to an absence of ovarian response, and age over 38 years increased the risk of miscarriage (sensitivity 61.5, specificity 73.8; AUC 0.705).
      Navali and co-workers also conducted a clinical study in 35 participants with POR (
      • Navali N
      • Sadeghi L
      • Farzadi L
      • Ghasemzadeh A
      • Hamdi K
      • Hakimi P
      • Niknafs B.
      Intraovarian Injection of Autologous Platelet-Rich Plasma Improves Therapeutic Approaches in The Patients with Poor Ovarian Response: A Before-After Study.
      ). After a 2-month follow up, they identified a significant increase in the number of oocytes and embryos obtained after IVF. Moreover, there was a significant increase in serum oestradiol concentrations after PRP treatment, while FSH and AMH concentrations presented no significant surge after a single injection of PRP. Three women reported a natural conception 4 months after the PRP treatment.
      A recent investigation suggests that PRP infusion did not increase ovarian reserve but instead had a positive effect on embryo euploidy rate (
      • Merhi Z
      • Seckin S
      • Mouanness M.
      Intraovarian platelet-rich plasma administration could improve blastocyst euploidy rates in women undergoing in vitro fertilization.
      ). This study included women with at least one previous failed IVF cycle and/or who produced blastocysts both before and after intra-ovarian PRP treatment. Although neither FSH, AMH nor AFC significantly improved after PRP, the euploidy rate increased from only 8% to 42.8% after PRP infusion. Similarly, a normalization of the embryonic chromosome content after PRP treatment was reported in a case study of a 42-year-old patient with six IVF previous attempts and a total of 20 aneuploid embryos (100% aneuploidy rate), who achieved an euploid embryo after PRP ovarian infusion followed 5 months later by an injection of isolated PDGF (
      • Sills ES
      • Rickers NS
      • Svid CS
      • Rickers JM
      • Wood SH.
      Normalized Ploidy Following 20 Consecutive Blastocysts with Chromosomal Error: Healthy 46, XY Pregnancy with IVF after Intraovarian Injection of Autologous Enriched Platelet-derived Growth Factors.
      ).
      An additional case report of a 35-year-old patient who suffered from long-term infertility and recurrent pregnancy loss (
      • Sabouni R
      • Tarrab R
      • Kalaji D
      • Abbassi H.
      A new approach of using platelet-rich autologous plasma to increase the ovarian reservoir in a Syrian patient with ovarian insufficiency: A case report.
      ) reported increased AMH concentrations (0.39 ng/ml versus 0.94 ng/ml) 15 days after PRP injections into different points of the reproductive system including the ovaries, cervix and uterus, and intramuscularly (the thighs).
      PRP treatment in women with POI is different because this group have amenorrhoea or oligomenorrhoea, high FSH concentrations and no antral follicles to be stimulated with exogenous gonadotrophins. A recent pilot study was performed in 311 women with POI who were less than 40 years age (
      • Cakiroglu Y
      • Saltik A
      • Yuceturk A
      • Karaosmanoglu O
      • Kopuk SY
      • Scott RT
      • Tiras B
      • Seli E.
      Effects of intraovarian injection of autologous platelet rich plasma on ovarian reserve and IVF outcome parameters in women with primary ovarian insufficiency.
      ) and in whom intra-ovarian PRP infusion into at least one ovary was performed. This investigation showed promising results, with a total pregnancy rate of 11.6%: 23 pregnancies were achieved naturally, while 13 were achieved after IVF (pregnancy rate of 22.8% per transfer, 4% of the total). Altogether, 8% accomplished a live birth, while another 8% were cryopreserving embryos at the time of publication. Interestingly, 201 participants showed follicular growth (64.6%) and were able to undergo ART. Additional case reports in POI also describe pregnancies (
      • Sfakianoudis K
      • Simopoulou M
      • Nitsos N
      • Rapani A
      • Pappas A
      • Pantou A
      • Chronopoulou M
      • Deligeoroglou E
      • Koutsilieris M
      • Pantos K.
      Autologous platelet-rich plasma treatment enables pregnancy for a woman in premature menopause.
      ) and live births (
      • Hsu CC
      • Hsu L
      • Hsu I
      • Chiu YJ
      • Dorjee S.
      Live Birth in Woman With Premature Ovarian Insufficiency Receiving Ovarian Administration of Platelet-Rich Plasma (PRP) in Combination With Gonadotropin: A Case Report.
      ) after PRP injection.
      Furthermore, PRP has been tested in women with perimenopause and menopause. The first pilot study (
      • Pantos K
      • Nitsos N
      • Kokkali G
      • Vaxevanoglou T
      • Markomichali C
      • Pantou A
      • Grammatis M
      • Lazaros L
      • Sfakianoudis K.
      Ovarian rejuvenation and folliculogenesis reactivation in peri-menopausal women after autologous platelet-rich plasma treatment.
      ), in eight perimenopausal women of advanced maternal age, showed a restoration of menstruation in all women 1–3 months after treatment, with oocyte retrieval and vitrification of embryos. Three years later, the same group described similar results in three women: one postmenopausal patient and two women with POI (
      • Pantos K
      • Simopoulou M
      • Pantou A
      • Rapani A
      • Tsioulou P
      • Nitsos N
      • Syrkos S
      • Pappas A
      • Koutsilieris M
      • Sfakianoudis K.
      A Case Series on Natural Conceptions Resulting in Ongoing Pregnancies in Menopausal and Prematurely Menopausal Women Following Platelet-Rich Plasma Treatment.
      ), all achieving a natural conception 2–6 months after PRP treatment along with restoration of menses, a decrease in FSH and an increase in AMH.
      Four different pilot studies in 120 patients with DOR, POI, perimenopause or menopause (30 with each pathology;
      • Sfakianoudis K
      • Simopoulou M
      • Grigoriadis S
      • Pantou A
      • Tsioulou P
      • Maziotis E
      • Rapani A
      • Giannelou P
      • Nitsos N
      • Kokkali G
      • Koutsilieris M
      • Pantos K.
      Reactivating ovarian function through autologous platelet-rich plasma intraovarian infusion: Pilot data on premature ovarian insufficiency, perimenopausal, menopausal, and poor responder women.
      ) showed that PRP infused on day 3 of the menstrual cycle, except in amenorrhoeic women, who received it on a random day, improved ovarian reserve biomarkers and the number of oocytes retrieved in women with DOR. In the first group, ovarian reserve biomarkers improved, especially in the first cycle after the intervention, and ICSI performance was enhanced after PRP treatment, with a 46.6% pregnancy rate (compared with no pregnancies before the intervention). In participants with POI, 60% of the women recruited had a restoration of their menstrual cycle, in addition to an improvement in hormone concentrations (AMH and FSH) and AFC. The pregnancy rate was 10%, and all were natural conceptions. In perimenopausal patients recovery of menses occurred in 80%, along with a reduction in FSH. The natural conception rate was 13.3%, followed by three live births. Restoration of menses was observed in 43.3% of the postmenopausal group, with hormone improvement and a 3.3% pregnancy rate with one live birth. The authors concluded that results were more optimistic in women with DOR and perimenopause than with POI and menopause.
      In summary, findings in patients with DOR show a restoration of ovarian function in the first 2–3 months after a PRP intervention (
      • Cakiroglu Y
      • Yuceturk A
      • Karaosmanoglu O
      • Kopuk SY
      • Korun ZEU
      • Herlihy N
      • Scott RT
      • Tiras B
      • Seli E.
      Ovarian reserve parameters and IVF outcomes in 510 women with poor ovarian response (POR) treated with intraovarian injection of autologous platelet rich plasma (PRP).
      ;
      • Melo P
      • Navarro C
      • Jones C
      • Coward K
      • Coleman L.
      The use of autologous platelet-rich plasma (PRP) versus no intervention in women with low ovarian reserve undergoing fertility treatment: a non-randomized interventional study.
      ;
      • Sabouni R
      • Tarrab R
      • Kalaji D
      • Abbassi H.
      A new approach of using platelet-rich autologous plasma to increase the ovarian reservoir in a Syrian patient with ovarian insufficiency: A case report.
      ;
      • Sills ES
      • Rickers NS
      • Li X
      • Palermo GD.
      First data on in vitro fertilization and blastocyst formation after intraovarian injection of calcium gluconate-activated autologous platelet rich plasma.
      ;
      • Tülek F
      • Kahraman A.
      The effects of intra-ovarian autologous platelet rich plasma injection on IVF outcomes of poor responder women and women with premature ovarian insufficiency.
      ), similar to that described after stem cell treatment. This is likely to be due to the rescue of secondary and pre-antral follicles, because the PRP stimulus prevents follicle atresia under normal conditions in poor-responder patients (
      • Melo P
      • Navarro C
      • Jones C
      • Coward K
      • Coleman L.
      The use of autologous platelet-rich plasma (PRP) versus no intervention in women with low ovarian reserve undergoing fertility treatment: a non-randomized interventional study.
      ;
      • Pellicer A
      • Ardiles G
      • Neuspiller F
      • Remohí J
      • Simón C
      • Bonilla-Musoles F.
      Evaluation of the ovarian reserve in young low responders with normal basal levels of follicle-stimulating hormone using three-dimensional ultrasonography.
      ). Follicle growth seen in both POI and perimenopausal or menopausal women is linked to primordial follicle stimulation. However, hormone improvement is not evident in all patients, and some still presented with amenorrha during the follow-up (
      • Sfakianoudis K
      • Simopoulou M
      • Grigoriadis S
      • Pantou A
      • Tsioulou P
      • Maziotis E
      • Rapani A
      • Giannelou P
      • Nitsos N
      • Kokkali G
      • Koutsilieris M
      • Pantos K.
      Reactivating ovarian function through autologous platelet-rich plasma intraovarian infusion: Pilot data on premature ovarian insufficiency, perimenopausal, menopausal, and poor responder women.
      ).
      Recent investigations have shown promising results (Table 3). However, the studies use different PRP volumes and activation protocols, and the population characteristics are heterogeneous, which is likely to lead to a variation in outcomes. This makes it difficult to obtain standardized protocols for future studies. Moreover, many of the investigations described are case reports with low-quality data, and controlled clinical trials are missing. Furthermore, as current clinical trials also lack a control group, some outcomes, such as hormone improvement or pregnancy rate after ART, may be inconclusive because some individuals conceive naturally while others opt to store cryopreserved embryos.
      Table 3Investigations assessing the effect of intra-ovarian PRP injection for follicular activation, according to the diagnoses of POR and POI, and in premenopausal and post-menopausal patients
      TechniqueDiagnosis and age of patient(s)Study designVolume injectedPlatelet activation methodOutcomes overall
      Bilateral activated PRP intra-ovarian infusion (
      • Sills ES
      • Rickers NS
      • Li X
      • Palermo GD.
      First data on in vitro fertilization and blastocyst formation after intraovarian injection of calcium gluconate-activated autologous platelet rich plasma.
      )
      DOR

      Mean age 42 ± 4 years
      Prospective clinical trial (n = 4) (NCT03178695)1 ml of activated PRP per ovaryCalcium gluconateNon-significant increase in serum AMH (P = 0.17) and decrease in FSH (P < 0.01) in all cases

      MII oocyte retrieval (5.3 ± 1.3 MII oocytes)

      At least one blastocyst per patient

      1 pregnancy (9 weeks gestational age)
      Bilateral PRP intra-ovarian infusion (
      • Farimani M
      • Heshmati S
      • Poorolajal J
      • Bahmanzadeh M.
      A report on three live births in women with poor ovarian response following intra-ovarian injection of platelet-rich plasma (PRP).
      )
      POR

      Age not specified
      Prospective clinical trial (n = 19) (IRCT201703079014N150)2 ml of PRP per ovaryNoOocyte yield increase from 0.6 to 2.1 after PRP infusion

      3 clinical pregnancies (2 natural conceptions)

      1 healthy live birth
      Bilateral activated PRP intra-ovarian infusion (
      • Melo P
      • Navarro C
      • Jones C
      • Coward K
      • Coleman L.
      The use of autologous platelet-rich plasma (PRP) versus no intervention in women with low ovarian reserve undergoing fertility treatment: a non-randomized interventional study.
      )
      POR

      Median age 41 years)
      Prospective controlled, non-randomized comparative study

      (n = 83)

      1. 46 patients with PRP infusion

      2. 37 patients with no intervention
      200 μl of activated PRP per ovary0.1 ml of 10% calcium chloride46/83 patients received the PRP infusion

      Significant FSH decrease (33%) and increase in AMH (63%) and AFC (75%) in the PRP group

      1.5 × more oocytes retrieved after IVF in the PRP group

      Higher rate of medium- and top-quality embryos in the PRP group (100% versus 55%)

      13 pregnancies in the treatment group versus 2 in the control group

      5 live births in PRP group, 2 natural conceptions, versus 1 in the control group after IVF
      Bilateral activated PRP intra-ovarian infusion (
      • Sfakianoudis K
      • Simopoulou M
      • Grigoriadis S
      • Pantou A
      • Tsioulou P
      • Maziotis E
      • Rapani A
      • Giannelou P
      • Nitsos N
      • Kokkali G
      • Koutsilieris M
      • Pantos K.
      Reactivating ovarian function through autologous platelet-rich plasma intraovarian infusion: Pilot data on premature ovarian insufficiency, perimenopausal, menopausal, and poor responder women.
      )
      POR

      Mean age 38 years
      Pilot study

      (n = 30)
      4 ml of PRP per ovaryCalcium gluconateImprovement of AMH and AFC on first menstrual cycle after PRP

      ICSI in 96.6% of patients

      101 oocytes retrieved, producing 58 embryos

      14 pregnancies with 2 miscarriages and 12 live births
      Bilateral PRP intra-ovarian infusion (
      • Farimani M
      • Nazari A
      • Mohammadi S
      • Anvari Aliabad R
      Evaluation of intra-ovarian platelet-rich plasma administration on oocytes-dependent variables in patients with poor ovarian response: A retrospective study according to the POSEIDON criteria.
      )
      POR

      Mean age 38.3 ± 4.5 years
      Retrospective study

      (n = 96)
      2 ml of PRP per ovaryNoNo statistical differences in hormone levels after treatment

      Increase in MII oocytes retrieved in POSEIDON groups 1, 3 and 4

      14 clinical pregnancies, higher pregnancy rate in POSEIDON group 1
      Bilateral PRP intra-ovarian infusion in 18 patients.

      In 2 patients, infusion during a laparoscopic intervention

      (
      • Pacu I
      • Zygouropoulos N
      • Dimitriu M
      • Rosu G
      • Ionescu CA.
      Use of platelet-rich plasma in the treatment of infertility in poor responders in assisted human reproduction procedures.
      )
      POR

      Mean age 37.4 years (31–44)
      Retrospective analysis (n = 20)2–4 ml per ovaryNoAMH and AFC increased in the first and second menstrual cycles after intervention

      IVF cancellation rate was decreased from 40% before to 15% after PRP

      Increase in the number of oocytes and embryos obtained after PRP versus previous IVF cycles

      2 pregnancies with 2 live births, 1 pregnancy being a natural conception
      Bilateral PRP intra-ovarian infusion (
      • Cakiroglu Y
      • Yuceturk A
      • Karaosmanoglu O
      • Kopuk SY
      • Korun ZEU
      • Herlihy N
      • Scott RT
      • Tiras B
      • Seli E.
      Ovarian reserve parameters and IVF outcomes in 510 women with poor ovarian response (POR) treated with intraovarian injection of autologous platelet rich plasma (PRP).
      )
      POR according to POSEIDON criteria

      Mean age 40.3 years (30–45)
      Prospective observational study

      (n = 510)
      PRP injected in both ovaries (dose not reported)NoStatistically significant increase in AFC (4.2 ± 2.4 versus 2.6 ± 1.3)

      105 pregnancies: 22 natural conceptions and 87 pregnancies after IVF

      66 live births, 54 after IVF and 12 from natural conception
      Bilateral PRP intra-ovarian infusion (
      • Barad DH
      • Albertini DF
      • Molinari E
      • Gleicher N.
      Preliminary report of intraovarian injections of autologous platelet-rich plasma (PRP) in extremely poor prognosis patients with only oocyte donation as alternative: a prospective cohort study.
      )
      Patients with poor prognosis after failed IVF, 54 with regular menses, 26 with oligomenorrhoeaMean age 44.2 ± 5.4 yearsProspective cohort study (n = 80)1.5 ml of PRP per ovaryNoNo amelioration of hormone concentrations (oestradiol, FSH or AMH) after PRP treatment

      6 clinical pregnancies after IVF, with 2 ongoing pregnancies and 2 live births
      Bilateral PRP intra-ovarian infusion (
      • Navali N
      • Sadeghi L
      • Farzadi L
      • Ghasemzadeh A
      • Hamdi K
      • Hakimi P
      • Niknafs B.
      Intraovarian Injection of Autologous Platelet-Rich Plasma Improves Therapeutic Approaches in The Patients with Poor Ovarian Response: A Before-After Study.
      )
      POR

      Mean age 40.43 ± 0.26 years
      Clinical trial study

      (n = 35)
      PRP injected into the cortex of both ovariesNoAfter a 2-month follow-up, patients displayed a significant elevation in the number of oocytes (3.68 ± 0.24 versus 2.22 ± 0.13) and embryos obtained (3.17 ± 0.14 versus 1.41 ± 0.13) compared with previous levels

      Significant increase in oestradiol levels (404.1 ± 16.76 versus 237.7 ± 13.14 pg/ml)

      3 spontaneous pregnancies (4 months after PRP treatment)
      Bilateral PRP intra-ovarian infusion (Tülek et al., 2022)POR

      Mean age 38.1 ± 4.4 years
      Retrospective analysis

      (n = 50)
      2 ml of PRP per ovaryNoSignificant increase in number of oocytes and embryos obtained following IVF

      Cancellation rate lower following PRP

      7 clinical pregnancies and 4 live births
      Bilateral PRP intra-ovarian infusion (
      • Merhi Z
      • Seckin S
      • Mouanness M.
      Intraovarian platelet-rich plasma administration could improve blastocyst euploidy rates in women undergoing in vitro fertilization.
      )
      Women with at least one failed IVF treatment

      Mean age 40.08 ± 1.46 years
      Prospective pilot study (n = 12)4 ml of PRP per ovaryNoNo significant changes in FSH and AMH levels, or AFC value after PRP

      Before PRP 3/37 embryos analysed were euploid, after PRP 12/28 embryos analysed were euploid
      Bilateral PRP intra-ovarian infusion – 5 months later followed by an injection of isolated platelet-derived growth factors prepared as an enriched, cell-free substrate (Sills et al, 2019)Advanced age – low ovarian reserve (42 years)Case report

      (n = 1)
      Not reportedNo1 euploid embryo after treatment

      20 aneuploid embryos prior to treatment
      PRP injection into different points of the reproductive organs via laparoscopy (
      • Sabouni R
      • Tarrab R
      • Kalaji D
      • Abbassi H.
      A new approach of using platelet-rich autologous plasma to increase the ovarian reservoir in a Syrian patient with ovarian insufficiency: A case report.
      5 years’ infertility

      35 years old
      Case report (n = 1)20 ml in total: 0.5 ml per ovary, 2 ml in the cervix, 7 ml intrauterine, 7 ml intramuscularly (thighs)NoAMH improvement from 0.39 to 0.94 n
      g/ml 15 days after the intervention
      Bilateral PRP intra-ovarian infusion (
      • Sfakianoudis K
      • Simopoulou M
      • Nitsos N
      • Rapani A
      • Pappas A
      • Pantou A
      • Chronopoulou M
      • Deligeoroglou E
      • Koutsilieris M
      • Pantos K.
      Autologous platelet-rich plasma treatment enables pregnancy for a woman in premature menopause.
      )
      POI

      40 years old
      Case report (n = 1)4 ml of PRP per ovaryNot reportedMenstrual cycle 6 weeks after PRP, with:

      1. AMH improvement (from 0.02 to 0.08 ng/ml)

      2. FSH decrease (from 149 to 27 mIU/ml)

      1 pregnancy after a natural cycle, with spontaneous miscarriage in week 5
      Bilateral PRP intra-ovarian infusion, combined with recombinant 150 IU of FSH + 75 IU LH (Pergoveris) (
      • Hsu CC
      • Hsu L
      • Hsu I
      • Chiu YJ
      • Dorjee S.
      Live Birth in Woman With Premature Ovarian Insufficiency Receiving Ovarian Administration of Platelet-Rich Plasma (PRP) in Combination With Gonadotropin: A Case Report.
      )
      POI

      37 years old
      Case report (n = 1)3 ml per ovary (with 1 ml of recombinant 150 IU of FSH and 75 IU of LH)Calcium gluconate2 IVF cycles performed in the first two menstrual cycles

      6 MII oocytes retrieved

      2 embryos obtained

      DET

      Twin pregnancy with 2 live births
      Bilateral PRP intra-ovarian infusion (
      • Pantos K
      • Simopoulou M
      • Pantou A
      • Rapani A
      • Tsioulou P
      • Nitsos N
      • Syrkos S
      • Pappas A
      • Koutsilieris M
      • Sfakianoudis K.
      A Case Series on Natural Conceptions Resulting in Ongoing Pregnancies in Menopausal and Prematurely Menopausal Women Following Platelet-Rich Plasma Treatment.
      )
      POI

      27 and 40 years old
      Case report (n = 2)4 ml of PRP per ovaryYes, but substance not reportedRestoration of menses

      FSH decrease:

      First patient: from 65 to 10 mIU/ml in the first menstrual cycle, 2 months following PRP

      Second patient: from 46.5 to 15.0 mIU/ml in the 5th month

      AMH increase

      2 naturally conceived ongoing pregnancies
      Bilateral or unilateral PRP intra-ovarian infusion (
      • Cakiroglu Y
      • Saltik A
      • Yuceturk A
      • Karaosmanoglu O
      • Kopuk SY
      • Scott RT
      • Tiras B
      • Seli E.
      Effects of intraovarian injection of autologous platelet rich plasma on ovarian reserve and IVF outcome parameters in women with primary ovarian insufficiency.
      )
      POI

      <40 years old
      Pilot study (n = 311)2–4 ml of PRPNot reported201 patients attempted IVF, and oocyte retrieval was performed in 130

      82 women obtained at least one cleavage-stage embryo

      57 patients underwent ET (for the remaining 25 patients embryos were vitrified)

      36 pregnancies, 23 natural conceptions and 13 conceptions after IVF.

      9 live births at the time of publication
      Bilateral PRP intra-ovarian infusion (
      • Sfakianoudis K
      • Simopoulou M
      • Grigoriadis S
      • Pantou A
      • Tsioulou P
      • Maziotis E
      • Rapani A
      • Giannelou P
      • Nitsos N
      • Kokkali G
      • Koutsilieris M
      • Pantos K.
      Reactivating ovarian function through autologous platelet-rich plasma intraovarian infusion: Pilot data on premature ovarian insufficiency, perimenopausal, menopausal, and poor responder women.
      )
      POI

      <40 years old
      Pilot study (n = 30)4 ml of PRP per ovaryCalcium gluconateRestoration of menstrual cycles in 18 of the 30 POI patients (success group): this subgroup presented a statistically significant improvement in hormone levels (AMH and FSH) and AFC

      3 natural conceptions with 3 live births
      Bilateral PRP intra-ovarian infusion (Tülek et al.,2022)POI

      Mean age 37.9 ± 1.9 years
      Retrospective analysis (n = 21)2 ml of PRP per ovaryNot reportedMenses restored in 10 patients

      8 embryos obtained and transferred, but no pregnancy
      Bilateral PRP intra-ovarian infusion (
      • Sfakianoudis K
      • Simopoulou M
      • Grigoriadis S
      • Pantou A
      • Tsioulou P
      • Maziotis E
      • Rapani A
      • Giannelou P
      • Nitsos N
      • Kokkali G
      • Koutsilieris M
      • Pantos K.
      Reactivating ovarian function through autologous platelet-rich plasma intraovarian infusion: Pilot data on premature ovarian insufficiency, perimenopausal, menopausal, and poor responder women.
      )
      Premenopausal

      ≥40 years old
      Pilot study

      (n = 30)
      4 ml of PRP per ovaryCalcium gluconateRestoration of menstrual cycle in 24 of the 30 women

      FSH reduction

      4 natural conceptions with 3 live births and 1 miscarriage
      PRP surrounding the ovarian graft (Callejo et al., 2013)Post-menopausal

      30 years old
      Case study (n = 1)Not reportedNot reportedRestoration of menses

      1 pregnancy

      1 live birth
      Bilateral PRP intra-ovarian infusion (
      • Pantos K
      • Nitsos N
      • Kokkali G
      • Vaxevanoglou T
      • Markomichali C
      • Pantou A
      • Grammatis M
      • Lazaros L
      • Sfakianoudis K.
      Ovarian rejuvenation and folliculogenesis reactivation in peri-menopausal women after autologous platelet-rich plasma treatment.
      )
      Post-menopausal

      Mean age 45.1 ± 4.4 years
      Case reports (n = 8)Not reportedNot reportedRestoration of menses in all 8 women

      2.5. ± 0.7 follicles observed

      1.5 ± 0.7 MII oocytes retrieved

      1.5 ± 0.7 embryos vitrified
      Bilateral PRP intra-ovarian infusion (
      • Pantos K
      • Simopoulou M
      • Pantou A
      • Rapani A
      • Tsioulou P
      • Nitsos N
      • Syrkos S
      • Pappas A
      • Koutsilieris M
      • Sfakianoudis K.
      A Case Series on Natural Conceptions Resulting in Ongoing Pregnancies in Menopausal and Prematurely Menopausal Women Following Platelet-Rich Plasma Treatment.
      )
      Post-menopausal

      46 years old
      Case reports (n = 1)4 ml of PRP per ovaryYes, but substance not reportedRestoration of regular menses since the first month after treatment

      FSH decrease (from 119 to 27 mIU/ml in the fifth month after PRP)

      AMH and AFC increased

      1 naturally conceived ongoing pregnancy
      Bilateral PRP intra-ovarian infusion (
      • Sfakianoudis K
      • Simopoulou M
      • Grigoriadis S
      • Pantou A
      • Tsioulou P
      • Maziotis E
      • Rapani A
      • Giannelou P
      • Nitsos N
      • Kokkali G
      • Koutsilieris M
      • Pantos K.
      Reactivating ovarian function through autologous platelet-rich plasma intraovarian infusion: Pilot data on premature ovarian insufficiency, perimenopausal, menopausal, and poor responder women.
      )
      Post-menopausal

      45–55 years old
      Pilot study (n = 30)4 ml of PRP per ovaryCalcium gluconateRestoration of menses was observed in 13 of 30 patients (success subgroup): in these patients there was statistical significant hormone improvement (AMH, FSH, LH)

      1 natural conception with 1 live birth
      AFC, antral follicle count; AMH, anti-Müllerian hormone, DET, double-embryo transfer; DOR, diminished ovarian reserve; ET, embryo transfer; ICSI, intracytoplasmic sperm injection; MII, metaphase II; POI, premature ovarian insufficiency; POR, poor ovarian response; POSEIDON, Patient-Oriented Strategies Encompassing IndividualizeD Oocyte Number; PRP, platelet-rich plasma.
      However, the conclusion that PRP treatment could be an alternative treatment for women with DOR is viable based on the increased pregnancy rates reported in the literature even though randomized controlled trials are lacking. Whereas the results are more encouraging in women with patients, optimistic outcomes were also obtained in those with POI and established menopause, whose reproductive prognosis is worse than that of DOR patients. In the future, PRP as an alternative therapy should be further investigated. There are several ongoing trials involving PRP injection to treat impaired ovaries (NCT04797377, NCT04275700, NCT04149028, NCT03542708 and NCT02992756), some of which already include a proper control group (NCT03937661, NCT05181748, NCT04922398 and NCT04381299), while others aim to compare PRP with PPP (NCT04278313) and another is combining PRP with stem cells (NCT04444245).

      Future insights into the mechanisms governing follicular dynamics and oocyte ageing

      Ovarian reactivation techniques focus largely on strengthening the process of follicle activation and growth. However, none has shown a relevant effect on increasing oocyte quality, and they rather assess aneuploidy rate by preimplantation genetic testing for aneuploidies, showing values according to patient age (
      • Cakiroglu Y
      • Yuceturk A
      • Karaosmanoglu O
      • Kopuk SY
      • Korun ZEU
      • Herlihy N
      • Scott RT
      • Tiras B
      • Seli E.
      Ovarian reserve parameters and IVF outcomes in 510 women with poor ovarian response (POR) treated with intraovarian injection of autologous platelet rich plasma (PRP).
      ;
      • Herraiz S
      • Romeu M
      • Buigues A
      • Martinez S
      • Diaz-Garcia C
      • Gomez-Segui I
      • Martinez J
      • Pellicer N
      • Pellicer A
      Autologous stem cell ovarian transplantation to increase reproductive potential in patients who are poor responders.
      ). Increasing follicle growth and final oocyte yield increases the possibility of having a good-quality oocyte to produce a euploid and transferable healthy embryo (
      • McCulloh DH
      • Alikani M
      • Norian J
      • Kolb B
      • Arbones JM
      • Munné S.
      Controlled ovarian hyperstimulation (COH) parameters associated with euploidy rates in donor oocytes.
      ). However, because the developmental potential of early embryos relies on oocyte quality, strategies to modify oocyte and embryo quality are increasingly requested as both parameters significantly decline with age (
      • Franasiak JM
      • Forman EJ
      • Hong KH
      • Werner MD
      • Upham KM
      • Treff NR
      • Scott Jr, RT
      The nature of aneuploidy with increasing age of the female partner: a review of 15,169 consecutive trophectoderm biopsies evaluated with comprehensive chromosomal screening.
      ).

      Maternal spindle transfer

      Aneuploidies in aged oocytes frequently result from impaired spindle assembly, one of the most energy-consuming phases of meiosis resumption (
      • Eichenlaub-Ritter U
      • Staubach N
      • Trapphoff T.
      Chromosomal and cytoplasmic context determines predisposition to maternal age-related aneuploidy: brief overview and update on MCAK in mammalian oocytes.
      ). Indeed, reduced mitochondrial content in aged oocytes also leads to impaired fertilization and embryo development (
      • Seifer DB
      • DeJesus V
      • Hubbard K.
      Mitochondrial deletions in luteinized granulosa cells as a function of age in women undergoing in vitro fertilization.
      ;
      • Suganuma N
      • Kitagawa T
      • Nawa A
      • Tomoda Y.
      Human ovarian aging and mitochondrial DNA deletion.
      ) because mitochondria provide the energy needed for these processes. However, developing a healthy embryo also requires input from other organelles and molecules within the ooplasm machinery that is impaired as age advances (
      • Bianchi S
      • Macchiarelli G
      • Micara G
      • Linari A
      • Boninsegna C
      • Aragona C
      • Rossi G
      • Cecconi S
      • Nottola SA
      Ultrastructural markers of quality are impaired in human metaphase II aged oocytes: a comparison between reproductive and in vitro aging.
      ;
      • Coticchio G
      • Dal Canto M
      • Mignini Renzini M
      • Guglielmo MC
      • Brambillasca F
      • Turchi D
      • Novara PV
      • Fadini R
      Oocyte maturation: gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization.
      ;
      • Hoffmann S
      • Król M
      • Polanski Z.
      Spindle assembly checkpoint-related meiotic defect in oocytes from LT/Sv mice has cytoplasmic origin and diminishes in older females.
      ;
      • Liu H
      • Wang CW
      • Grifo JA
      • Krey LC
      • Zhang J.
      Reconstruction of mouse oocytes by germinal vesicle transfer: maturity of host oocyte cytoplasm determines meiosis.
      ;
      • Reader KL
      • Stanton JL
      • Juengel JL
      The Role of Oocyte Organelles in Determining Developmental Competence.
      ;
      • Watson AJ.
      Oocyte cytoplasmic maturation: a key mediator of oocyte and embryo developmental competence.
      ).
      To overcome this, maternal spindle transfer (MST) was developed in a preclinical study and set up in human oocytes to provide the appropriate cytoplasmic and mitochondrial machinery from a donor to properly develop a euploid embryo using oocytes from different sources with minimal heteroplasmy (
      • Costa-Borges N.
      Spindle transfer can enhance the potential of developmentally compromised human oocytes to reach the blastocyst stage: proof of concept with donor oocytes. 34rd Annual Meeting of the European Society of Human Reproduction and Embryology.
      ;
      • Costa-Borges N
      • Spath K
      • Miguel-Escalada I
      • Mestres E
      • Balmaseda R
      • Serafín A
      • Garcia-Jiménez M
      • Vanrell I
      • González J
      • Rink K
      • Wells D
      • Calderón G.
      Maternal spindle transfer overcomes embryo developmental arrest caused by ooplasmic defects in mice.
      ). MST was successfully used in >90% of reconstructed oocytes (
      • Costa-Borges N
      • Nikitos E
      • Spath K
      • Rink K
      • Kostaras K
      • Zervomanolakis I
      • Kontopoulos G
      • Polyzos P
      • Grigorakis S
      • Prokopakis T
      • Vasilopoulos Y.
      • Vlahos N.
      • de Ziegler D.
      • Wells D.
      • Psathas P.
      • Calderon G.
      First registered pilot trial to validate the safety and effectiveness of maternal spindle transfer to overcome infertility associated with poor oocyte quality.
      ) in a trial with human oocytes from 25 donors with previous massive embryo arrest, leading to increased fertilization rates after ICSI and to the production of good-quality blastocysts (60.5%) with an overall 50% euploidy rate and less than 1% mitochondrial DNA carryover in the offspring. MST produced at least one good-quality blastocyst in 65% of women, allowing nine embryo transfers and resulting in six clinical pregnancies leading to two live births (and three ongoing pregnancies) in a very poor-prognosis population due to embryo arrest (
      • Costa-Borges N
      • Nikitos E
      • Spath K
      • Rink K
      • Kostaras K
      • Zervomanolakis I
      • Kontopoulos G
      • Polyzos P
      • Grigorakis S
      • Prokopakis T
      • Vasilopoulos Y.
      • Vlahos N.
      • de Ziegler D.
      • Wells D.
      • Psathas P.
      • Calderon G.
      First registered pilot trial to validate the safety and effectiveness of maternal spindle transfer to overcome infertility associated with poor oocyte quality.
      ).

      DNA damage response and repair

      Recent reports have highlighted the involvement of DNA damage response and repair, metabolism and apoptosis-regulating pathways and genes in the control of ovarian ageing (
      • Ruth KS
      • Day FR
      • Hussain J
      • Martínez-Marchal A
      • Aiken CE
      • Azad A
      • Thompson DJ
      • Knoblochova L
      • Abe H
      • Tarry-Adkins JL
      • et al.
      Genetic insights into biological mechanisms governing human ovarian ageing.
      ), supporting the fact that a variation in cell death after DNA damage response is a crucial mechanism in this process (Figure 3). The competence of follicular cells and oocytes to respond to DNA insults is critical to ensure their viability. In fact, prophase-arrested oocytes, especially those enclosed in dormant primordial follicles, retain the ability to repair exogenous damage through homologous recombination to ensure the transmission of an intact genome if apoptosis activation allows it (
      • Stringer JM
      • Winship A.
      Oocytes can efficiently repair DNA double-strand breaks to restore genetic integrity and protect offspring health.
      ).
      Figure 3
      Figure 3Molecular pathways related to oocyte quality with translational potential to extend the ovarian lifespan. Oocyte quality is affected by several factors such as age, disease, impaired bioenergetics, oxidative and metabolic stress, and exogenous agents. Different approaches have been proposed to recover oocyte quality: restoring telomere length and telomerase activity with pharmacological treatments; balancing metabolic and oxidative stress in early-stage oocytes through nicotinamide adenine dinucleotide (NAD+) repletion; improving mitochondrial function and supplementing the oocyte with autologous good-quality mitochondria; maternal spindle transfer to provide the oocyte with an adequate cytoplasmic and mitochondrial machinery from a donor to properly develop a euploid embryo; and priming the DNA damage and repair mechanisms to avoid follicle atresia induced by double-strand breaks by lowering CHEK2 activity to increase the time frame for DNA repair by homologous recombination when possible. Created with BioRender.com.
      This pathway involves a conserved DNA damage response checkpoint kinase ATM–CHEK2 (ataxia-telangiectasia mutated–checkpoint serine-threonine kinase 2). The role of CHEK2 is to integrate and determine repair and cellular responses from several DNA repair pathways and to promote apoptosis in response to double-strand breaks (
      • Bolcun-Filas E
      • Rinaldi VD
      • White ME
      • Schimenti JC.
      Reversal of female infertility by Chk2 ablation reveals the oocyte DNA damage checkpoint pathway.
      ), the most deleterious form of DNA damage inducing chromosome rearrangements and mutations (
      • Titus S
      • Li F
      • Stobezki R
      • Akula K
      • Unsal E
      • Jeong K
      • Dickler M
      • Robson M
      • Moy F
      • Goswami S
      • Oktay K.
      Impairment of BRCA1-related DNA double-strand break repair leads to ovarian aging in mice and humans.
      ). CHEK2 reduces the threshold of apoptosis activation in damaged primordial oocytes, allowing DNA repair and therefore reducing follicular depletion. Indeed, carriers of CHEK2 loss-of-function variants showed natural menopause 3.5 years later than non-carriers (
      • Ruth KS
      • Day FR
      • Hussain J
      • Martínez-Marchal A
      • Aiken CE
      • Azad A
      • Thompson DJ
      • Knoblochova L
      • Abe H
      • Tarry-Adkins JL
      • et al.
      Genetic insights into biological mechanisms governing human ovarian ageing.
      ), supporting the fact that reduced CHEK2 activity allows oocyte restoration and increases reproductive lifespan. Aged Chek2–/– knockout mice showed increased ovarian reserve, response to ovarian stimulation and reproductive potential even with a normal follicular reserve at birth (
      • Ruth KS
      • Day FR
      • Hussain J
      • Martínez-Marchal A
      • Aiken CE
      • Azad A
      • Thompson DJ
      • Knoblochova L
      • Abe H
      • Tarry-Adkins JL
      • et al.
      Genetic insights into biological mechanisms governing human ovarian ageing.
      ). This finding might be related to reduced follicular death and atresia as a consequence of lowered CHEK2 levels to allow the rescue of a small number of primordial follicles, suggesting a new strategy for future therapies.

      Oocyte mitochondrial bioenergetics

      Double-strand breaks are caused by exogenous genotoxic agents as well as by physiological oxidative and metabolic stress due to age (
      • Titus S
      • Li F
      • Stobezki R
      • Akula K
      • Unsal E
      • Jeong K
      • Dickler M
      • Robson M
      • Moy F
      • Goswami S
      • Oktay K.
      Impairment of BRCA1-related DNA double-strand break repair leads to ovarian aging in mice and humans.
      ), as recent studies using single-cell RNA sequencing have confirmed oxidative damage as the main age-associated threat to ovarian cells (
      • Wang S
      • Zheng Y
      • Li J
      • Yu Y
      • Zhang W
      • M
      • Liu Z
      • Min Z
      • Hu H
      • Jing Y
      • He X
      • Sun L
      • Ma L
      • Esteban CR
      • Chan P
      • Qiao J
      • Zhou Q
      • Izpisua Belmonte JC
      • Qu J
      • Tang F
      • Liu GH
      Single-Cell Transcriptomic Atlas of Primate Ovarian Aging.
      ) (Figure 3). Impaired mitochondrial bioenergetics, increased reactive oxygen species, DNA damage, apoptosis and spindle instabilities during chromosome segregation affect oocyte quality during ageing (
      • Franasiak JM
      • Forman EJ
      • Hong KH
      • Werner MD
      • Upham KM
      • Treff NR
      • Scott Jr, RT
      The nature of aneuploidy with increasing age of the female partner: a review of 15,169 consecutive trophectoderm biopsies evaluated with comprehensive chromosomal screening.
      ). Indeed, early-stage oocytes and granulosa cells from aged non-human primate ovaries and aged human granulosa cells showed a down-regulation of antioxidant genes such as IDH1, PRDX4 and NDUFB10, increased oxidative damage and apoptosis (
      • Wang S
      • Zheng Y
      • Li J
      • Yu Y
      • Zhang W
      • M
      • Liu Z
      • Min Z
      • Hu H
      • Jing Y
      • He X
      • Sun L
      • Ma L
      • Esteban CR
      • Chan P
      • Qiao J
      • Zhou Q
      • Izpisua Belmonte JC
      • Qu J
      • Tang F
      • Liu GH
      Single-Cell Transcriptomic Atlas of Primate Ovarian Aging.
      ). The observation that impaired bioenergetics and oxidative stress appeared in early-stage oocytes might explain the ineffectiveness of treatments intended to restore mitochondrial bioenergetic balance in mature oocytes during ICSI through the transfer of autologous mitochondria, as these treatments were applied when the damage has already been established in early development (
      • Labarta E
      • de Los Santos MJ
      • Herraiz S
      • Escribá MJ
      • Marzal A
      • Buigues A
      • Pellicer A
      Autologous mitochondrial transfer as a complementary technique to intracytoplasmic sperm injection to improve embryo quality in patients undergoing in vitro fertilization-a randomized pilot study.
      ).
      To amend treatment timing, a pharmacological intervention to restore metabolic and oxidative stress balance in early-stage oocytes was proposed based on the repletion of the redox cofactor nicotinamide adenine dinucleotide (NAD+/NADH), whose deficiency is related to several age-associated pathologies (
      • Hong W
      • Mo F
      • Zhang Z
      • Huang M
      • Wei X
      Nicotinamide Mononucleotide: A Promising Molecule for Therapy of Diverse Diseases by Targeting NAD+ Metabolism.
      ) and can be reversed by providing metabolic precursors such as nicotinamide mononucleotide (NMN) (
      • Bertoldo MJ
      • Listijono DR
      • Ho WJ
      • Riepsamen AH
      • Goss DM
      • Richani D
      • Jin XL
      • Mahbub S
      • Campbell JM
      • Habibalahi A
      • Loh WN
      • Youngson NA
      • Maniam J
      • Wong ASA
      • Selesniemi K
      • Bustamante S
      • Li C
      • Zhao Y
      • Marinova MB
      • Kim LJ
      • Lau L
      • Wu RM
      • Mikolaizak AS
      • Araki T
      • Le Couteur DG
      • Turner N
      • Morris MJ
      • Walters KA
      • Goldys E
      • O'Neill C
      • Gilchrist RB
      • Sinclair DA
      • Homer HA
      • Wu LE
      NAD(+) Repletion Rescues Female Fertility during Reproductive Aging.
      ). In the context of ovarian tissue, the age-associated decline in NAD+ is more prone to affect the oocyte rather than other ovarian cells. Oral administration of NMN for 4 weeks, a sufficient period for early-stage mouse follicles to activate, grow and mature (
      • Pedersen T
      • Peters H.
      Proposal for a classification of oocytes and follicles in the mouse ovary.
      ), improved final oocyte yield after ovarian stimulation, spindle assembly, blastocyst formation and euploidy rates in aged mice due to NAD+ repletion in oocytes (
      • Bertoldo MJ
      • Listijono DR
      • Ho WJ
      • Riepsamen AH
      • Goss DM
      • Richani D
      • Jin XL
      • Mahbub S
      • Campbell JM
      • Habibalahi A
      • Loh WN
      • Youngson NA
      • Maniam J
      • Wong ASA
      • Selesniemi K
      • Bustamante S
      • Li C
      • Zhao Y
      • Marinova MB
      • Kim LJ
      • Lau L
      • Wu RM
      • Mikolaizak AS
      • Araki T
      • Le Couteur DG
      • Turner N
      • Morris MJ
      • Walters KA
      • Goldys E
      • O'Neill C
      • Gilchrist RB
      • Sinclair DA
      • Homer HA
      • Wu LE
      NAD(+) Repletion Rescues Female Fertility during Reproductive Aging.
      ). These encouraging results were confirmed in breeding trials showing improved pregnancy rates of healthy offspring, highlighting the absence of deleterious effects associated with NMN supplementation. Moreover, the authors evaluated the role of NMN supplementation for in-vitro maturation and embryo culture and observed a restoration of oocyte quality.
      These findings, together with other reports supplementing several NAD precursors (
      • Guo Z
      • Yang J
      • Yang G
      • Feng T
      • Zhang X
      • Chen Y
      • Feng R
      • Qian Y.
      Effects of nicotinamide on follicular development and the quality of oocytes.
      ;
      • Pollard CL
      • Gibb Z
      • Clulow J
      • Ruiz A
      • Sheridan A
      • Bahrami M
      • Swegen A
      • Grupen CG.
      Supplemental Nicotinic Acid Elevates NAD+ Precursors in the Follicular Fluid of Mares.
      ), suggest a new approach to restoring the metabolic and oxidative balance of primordial follicles to obtain competent oocytes to produce implantable embryos for aged and young women with poor-quality embryos (Figure 3). Indeed, the ability of primordial follicles to remain dormant but viable and metabolically active for decades relies on their competence to limit reactive oxygen species production through the inhibition of the NADH dehydrogenase (complex I) of the respiratory chain (
      • Rodríguez-Nuevo A
      • Torres-Sanchez A
      • Duran JM
      • De Guirior C
      • Martínez-Zamora MA
      • Böke E
      Oocytes maintain ROS-free mitochondrial metabolism by suppressing complex I.
      ).

      Telomere maintenance and elongation

      Among the factors determining IVF success in ovarian and age-related infertility, telomeres are an attractive target. Telomeres are specialized structures at the ends of chromosomes whose main function is to preserve genome integrity and avoid chromosome fusions (
      • Martínez P.
      • Blasco M.A.
      Telomere-driven diseases and telomere-targeting therapies.
      ). Telomerase activity ensures the maintenance of telomere length by repairing the telomere shortening associated with each DNA replication cycle.
      During folliculogenesis, telomerase activity decreases and telomere length increases while primordial follicles advance to the antral stage (
      • Ozturk S.
      • Sozen B.
      • Demir N.
      Telomere length and telomerase activity during oocyte maturation and early embryo development in mammalian species.
      ), and telomerase activity then remains minimal during oocyte maturation and is reactivated at the blastocyst stage (
      • Wright D.L.
      • Jones E.L.
      • Mayer J.F.
      • Oehninger S.
      • Gibbons W.E.
      • Lanzendorf S.E.
      Characterization of telomerase activity in the human oocyte and preimplantation embryo.
      ). Interestingly, the oocyte and surrounding granulosa cells have shown differences in telomere length and telomerase activity, which is especially relevant in granulosa cells due to their impressive proliferation rate. Indeed, a compromised telomere pathway has been found in granulosa (
      • Butts S.
      • Riethman H.
      • Ratcliffe S.
      • Shaunik A.
      • Coutifaris C.
      • Barnhart K.
      Correlation of telomere length and telomerase activity with occult ovarian insufficiency.
      ;
      • Xu X.
      • Chen X.
      • Zhang X.
      • Liu Y.
      • Wang Z.
      • Wang P.
      • Du Y.
      • Qin Y.
      • Chen Z.J.
      Impaired telomere length and telomerase activity in peripheral blood leukocytes and granulosa cells in patients with biochemical primary ovarian insufficiency.
      ) and peripheral blood (
      • Xu X.
      • Chen X.
      • Zhang X.
      • Liu Y.
      • Wang Z.
      • Wang P.
      • Du Y.
      • Qin Y.
      • Chen Z.J.
      Impaired telomere length and telomerase activity in peripheral blood leukocytes and granulosa cells in patients with biochemical primary ovarian insufficiency.
      ) from young women with impaired ovarian function.
      Telomere-related parameters at the time of oocyte retrieval are predictive variables of IVF outcomes and aneuploidy. A higher relative telomere length has been detected in the surrounding cumulus cells of oocytes that fertilized and developed into a good-quality day 3 embryo (
      • Cheng EH
      • Chen SU
      • Lee TH
      • Pai YP
      • Huang LS
      • Huang CC
      • Lee MS
      Evaluation of telomere length in cumulus cells as a potential biomarker of oocyte and embryo quality.
      ) while telomerase activity in luteinized granulosa predicted clinical pregnancy in a cohort of young women (<38 years) (
      • Wang W
      • Chen H
      • Li R
      • Ouyang N
      • Chen J
      • Huang L
      • Mai M
      • Zhang N
      • Zhang Q
      • Yang D.
      Telomerase activity is more significant for predicting the outcome of IVF treatment than telomere length in granulosa cells.
      ). In the oocyte, short telomeres are associated with early embryo fragmentation and apoptosis (
      • Keefe D.L.
      • Franco S.
      • Liu L.
      • Trimarchi J.
      • Cao B.
      • Weitzen S.
      • Agarwal S.
      • Blasco M.A.
      Telomere length predicts embryo fragmentation after in vitro fertilization in women toward a telomere theory of reproductive aging in women.
      ) and aneuploidies (
      • Treff NR
      • Su J
      • Taylor D
      • Scott Jr., RT
      Telomere DNA deficiency is associated with development of human embryonic aneuploidy.
      ), while longer telomere length correlates with improved IVF pregnancy outcomes (
      • Keefe D.L.
      • Liu L.
      • Marquard K.
      Telomeres and aging-related meiotic dysfunction in women.
      ). Oocyte telomere function ensures chromosomal alignment and sustains spindle organization (
      • Liu L.
      • Blasco M.
      • Trimarchi J.
      • Keefe D.
      An essential role for functional telomeres in mouse germ cells during fertilization and early development.
      ;
      • Liu L.
      • Blasco M.A.
      • Keefe D.L.
      Requirement of functional telomeres for metaphase chromosome alignments and integrity of meiotic spindles.
      ), with telomere length being the crucial factor for both processes; this can be observed in telomerase-deficient knockout (Tr–/–) mice (
      • Liu L.
      • Blasco M.A.
      • Keefe D.L.
      Requirement of functional telomeres for metaphase chromosome alignments and integrity of meiotic spindles.
      ), in which breakpoints that resulted in telomere shortening in mature oocytes led to failed fertilization or the failed development of an implantable healthy embryo (
      • Liu L.
      • Blasco M.
      • Trimarchi J.
      • Keefe D.
      An essential role for functional telomeres in mouse germ cells during fertilization and early development.
      ).
      Pharmacological approaches to the treatment of short telomeres serve to activate telomerase activity in different ageing and degenerative diseases (
      • Fernandez M.L.
      • Thomas M.S.
      • Lemos B.S.
      • DiMarco D.M.
      • Missimer A.
      • Melough M.
      • Chun O.K.
      • Murillo A.G.
      • Alyousef H.M.
      • Medina-Vera I.
      TA-65, A Telomerase Activator improves Cardiovascular Markers in Patients with Metabolic Syndrome.
      ;
      • Harley C.B.
      • Liu W.
      • Blasco M.
      • Vera E.
      • Andrews W.H.
      • Briggs L.A.
      • Raffaele J.M.
      A natural product telomerase activator as part of a health maintenance program.
      ;
      • Harley C.B.
      • Liu W.
      • Flom P.L.
      • Raffaele J.M.
      A natural product telomerase activator as part of a health maintenance program: metabolic and cardiovascular response.
      ;
      • Hobbs T.
      • Caso R.
      • McMahon D.
      • Nymark M.
      A novel, multi-ingredient supplement to manage elevated blood lipids in patients with no evidence of cardiovascular disease: a pilot study.
      ;
      • Prieto-Oliveira P.
      Telomerase activation in the treatment of aging or degenerative diseases: a systematic review.
      ;
      • Salvador L.
      • Singaravelu G.
      • Harley C.B.
      • Flom P.
      • Suram A.
      • Raffaele J.M.
      A Natural Product Telomerase Activator Lengthens Telomeres in Humans: A Randomized, Double Blind, and Placebo Controlled Study.
      ;
      • Townsley D.M.
      • Dumitriu B.
      • Liu D.
      • Biancotto A.
      • Weinstein B.
      • Chen C.
      • Hardy N.
      • Mihalek A.D.
      • Lingala S.
      • Kim Y.J.
      • Yao J.
      • Jones E.
      • Gochuico B.R.
      • Heller T.
      • Wu C.O.
      • Calado R.T.
      • Scheinberg P.
      • Young N.S.
      Danazol Treatment for Telomere Diseases.
      ;
      • Tsoukalas D
      • Buga AM
      • Docea AO
      • Sarandi E
      • Mitrut R
      • Renieri E
      • Spandidos DA
      • Rogoveanu I
      • Cercelaru L
      • Niculescu M
      • Tsatsakis A
      • Calina D.
      Reversal of brain aging by targeting telomerase: A nutraceutical approach.
      ). Telomere-lengthening capability relies on the oocyte, as demonstrated by the telomere elongation observed in parthenogenetically activated oocytes (
      • Liu L
      • Bailey SM
      • Okuka M
      • Muñoz P
      • Li C
      • Zhou L
      • Wu C
      • Czerwiec E
      • Sandler L
      • Seyfang A
      • Blasco MA
      • Keefe DL.
      Telomere lengthening early in development.
      ). Thus, follicular cells can be considered as a potential target for this type of therapy, which promotes telomere lengthening in order to improve the outcomes of impaired and aged oocytes (Figure 3).

      Plasma- and growth factor-based therapies to restore folliculogenesis and oocyte quality

      The human ovary shows senescence and ageing signs early in life compared with other organs (Richardson et al., 2014;
      • Wang S
      • Zheng Y
      • Li J
      • Yu Y
      • Zhang W
      • M
      • Liu Z
      • Min Z
      • Hu H
      • Jing Y
      • He X
      • Sun L
      • Ma L
      • Esteban CR
      • Chan P
      • Qiao J
      • Zhou Q
      • Izpisua Belmonte JC
      • Qu J
      • Tang F
      • Liu GH
      Single-Cell Transcriptomic Atlas of Primate Ovarian Aging.
      ), with a sudden decrease of ovarian reserve after the mid-30s. Although systemic ageing effects seem to manifest around a woman's 60s, a recent characterization of the human plasma proteome identified three different waves of ageing defined by significant changes in protein composition at 34, 60 and 78 years of age (
      • Lehallier B
      • Gate D
      • Schaum N
      • Nanasi T
      • Lee SE
      • Yousef H
      • Moran Losada P
      • Berdnik D
      • Keller A
      • Verghese J
      • Sathyan S
      • Franceschi C
      • Milman S
      • Barzilai N
      • Wyss-Coray T
      Undulating changes in human plasma proteome profiles across the lifespan.
      ). Proteomic changes observed around 34 years of age involved a higher number of regulated proteins when compared with those occurring at 60 years and therefore could be involved, or at least be connected, with the early onset of ovarian ageing. Thus, the use of ‘omics’ analyses in ageing studies provides new and useful insights for anti-ageing therapies.
      Induction of tissue repair by introducing young growth factor-enriched plasma into damaged and aged organisms (
      • Castellano JM
      • Kirby ED
      • Wyss-Coray T
      Blood-Borne Revitalization of the Aged Brain.
      ;
      • Iram T
      • Kern F
      • Kaur A
      • Myneni S
      • Morningstar AR
      • Shin H
      • Garcia MA
      • Yerra L
      • Palovics R
      • Yang AC
      • Hahn O
      • Lu N
      • Shuken SR
      • Haney MS
      • Lehallier B
      • Iyer M
      • Luo J
      • Zetterberg H
      • Keller A
      • Zuchero JB
      • Young Wyss-Coray T.
      CSF restores oligodendrogenesis and memory in aged mice via Fgf17.
      ;
      • Villeda SA
      • Wyss-Coray T.
      The circulatory systemic environment as a modulator of neurogenesis and brain aging.
      ;
      • Villeda SA
      • Plambeck KE
      • Middeldorp J
      • Castellano JM
      • Mosher KI
      • Luo J
      • Smith LK
      • Bieri G
      • Lin K
      • Berdnik D
      • Wabl R
      • Udeochu J
      • Wheatley EG
      • Zou B
      • Simmons DA
      • Xie XS
      • Longo FM
      • Wyss-Coray T
      Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice.
      ) suggests that tissue damage and ageing could be in part attributed to changes in blood-borne factors (
      • Villeda SA
      • Luo J
      • Mosher KI
      • Zou B
      • Britschgi M
      • Bieri G
      • Stan TM
      • Fainberg N
      • Ding Z
      • Eggel A
      • Lucin KM
      • Czirr E
      • Park JS
      • Couillard-Després S
      • Aigner L
      • Li G
      • Peskind ER
      • Kaye JA
      • Quinn JF
      • Galasko DR
      • Xie XS
      • Rando TA
      • Wyss-Coray T.
      The ageing systemic milieu negatively regulates neurogenesis and cognitive function.
      ). The current authors’ recent work seeking to evaluate the potential of paracrine signalling of different stem cell sources in combination with additional platelet-enclosed factors showed that this can be a feasible and less invasive option to reactivate ovarian function (
      • Buigues A
      • Marchante M
      • de Miguel-Gómez L
      • Martinez J
      • Cervelló I
      • Pellicer A
      • Herraiz S.
      Stem cell-secreted factor therapy regenerates the ovarian niche and rescues follicles.
      ). Although plasma treatments enriched in BMDSC or umbilical cord stem cells reactivated follicle growth in POI mouse models, BMSDC had a more potent effect in recovering oocyte quality and short- and long-term fertility.
      Moreover, the activation process to release additional factors enclosed within platelets increased the effects, which were especially relevant in BMDSC-rich plasma. This difference might be due to a variation in the platelet secretome according to platelet origin (
      • Dregalla RC
      • Herrera JA
      • Donner EJ.
      Soluble factors differ in platelets derived from separate niches: a pilot study comparing the secretome of peripheral blood and bone marrow platelets.
      ). These results were also validated in human ovarian biopsies of women with POR xenografted into immunodeficient mice, which is currently being evaluated in an ongoing randomized clinical trial of women with POI that includes a non-treated control group (NCT04475744). The proteomic analysis of this plasma revealed the presence of soluble factors related to cell cycle/apoptosis, gene expression, signal transduction, cell communication, response to stress, signalling by RhoGTPases, nuclear receptors, NOTCH (notch receptor), PDGF and WNT (Wnt family member), and DNA repair of DBS (
      • Buigues A
      • Marchante M
      • de Miguel-Gómez L
      • Martinez J
      • Cervelló I
      • Pellicer A
      • Herraiz S.
      Stem cell-secreted factor therapy regenerates the ovarian niche and rescues follicles.
      ) induced by age and cancer treatments in oocytes (
      • Winship AL
      • Stringer JM
      • Liew SH
      • Hutt KJ.
      The importance of DNA repair for maintaining oocyte quality in response to anti-cancer treatments, environmental toxins and maternal ageing.
      ). These findings may be relevant to age-induced ovarian damage in addition to chemotherapy insults as a link between reduced oxidant stress response, cell communication, metabolism of proteins and ovarian ageing was recently described (
      • Wang S
      • Zheng Y
      • Li J
      • Yu Y
      • Zhang W
      • M
      • Liu Z
      • Min Z
      • Hu H
      • Jing Y
      • He X
      • Sun L
      • Ma L
      • Esteban CR
      • Chan P
      • Qiao J
      • Zhou Q
      • Izpisua Belmonte JC
      • Qu J
      • Tang F
      • Liu GH
      Single-Cell Transcriptomic Atlas of Primate Ovarian Aging.
      ).
      While these approaches are based on the rescue, repair and reactivation of existing follicles in damaged or senescent ovaries following senotherapeutic interventions (
      • Secomandi L
      • Borghesan M
      • Velarde M
      • Demaria M.
      The role of cellular senescence in female reproductive aging and the potential for senotherapeutic interventions.
      ), there are cases where access to germ cells is not an option and therefore a completely different approach should be taken.

      De-novo generation of oocytes

      In recent decades, differentiation of stem cell types into oocytes has been a challenge. Nevertheless, a series of cutting-edge studies to differentiate mouse embryonic and induced pluripotent stem cells into fertilizable oocytes (
      • Hayashi K
      • Ohta H
      • Kurimoto K
      • Aramaki S
      • Saitou M.
      Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells.