ArticlePDF Available

Primary Pulmonary Metastatic Osteosarcoma: To Stop or Not to Stop, That is the Question

Authors:

Abstract

An 18-year-old presented with a large, high-grade osteosarcoma of the proximal femur and multiple primary pulmonary metastases. The tumor responded poorly to preoperative chemotherapy. Four thoracotomies to remove 24 metastases were necessary to achieve a first complete remission. An early pulmonary recurrence was again treated by thoracotomy and systemic therapy. The patient remained in second complete remission at the last follow-up, 76 months after post-diagnosis and 53 months after pulmonary recurrence. This case illustrates that some patients with osteosarcoma and extremely doubtful prognosis may be cured, provided the treatment concept includes aggressive surgery for both the primary tumor and metastatic disease.
Perspective
Cryopreservation and Autotransplantation
of Ovarian Tissue in Cancer Patients: Is It Safe?
Lobke Bastings, MD, Catharina C.M. Beerendonk, MD, PhD,
Johan R. Westphal, PhD, Didi D.M. Braat, MD, PhD, and Ron Peek, PhD
Keywords: counseling, cryopreservation, autotransplantation, ovarian tissue, fertility preservation, safety
Oncological therapy may severely compromise the
future fertility of girls and young women with cancer
and thereby limit their quality of life. Various strategies to
preserve fertility before the start of gonadotoxic treatment
have been proposed, such as in vitro fertilization (IVF), in-
tracytoplasmic sperm injection (ICSI), vitrification of oocytes,
and cryopreservation of ovarian tissue.
1
Unfortunately, not
all of these techniques are suitable for pre-pubertal girls and
adolescents. Not only do IVF and ICSI require a male partner
or donor sperm for fertilization, procedures using gonado-
trophin administration and oocyte retrieval are considered
inappropriate for sexually immature patients.
2
In case of ovarian tissue cryopreservation, neither
hormonal stimulation nor a stable relationship is necessary.
These characteristics make ovarian tissue cryopreservation
especially suitable for adolescents and the only option
available for fertility preservation in pre-pubertal girls.
1,2
Furthermore, as ovarian tissue can be obtained directly after
diagnosis, there is only minimal interference with cancer
treatment.
1
The main aim of cryopreservation of ovarian tissue is to
restore reproductive potential by retransplanting the tissue.
This can be performed once the patient has overcome her
disease and wishes to conceive but experiences premature
ovarian insufficiency due to the cancer treatment.
1,3
Although
the technique of ovarian tissue autotransplantation and sub-
sequent autografting is still considered experimental, 16 live
births have already been reported; 12 of these children were
born to 9 women who survived cancer.
3,4
As no central registration for ovarian tissue cryopreserva-
tion or autotransplantation exists, the total number of pro-
cedures carried out worldwide is unknown.
5
Nevertheless,
a considerable number of cryopreservation and autotrans-
plantation procedures have been reported,
6–14
indicating that
cryopreservation of ovarian tissue have been performed
on a large scale in the past decade. Therefore, the number
of cancer survivors requesting autotransplantation of their
ovarian tissue is anticipated to increase considerably in the
near future.
5
Although autotransplantation procedures are already be-
ing performed in oncological patients worldwide, the risks of
recurrent malignancy due to transmission of cancer cells via
the ovarian graft still remains largely unknown. This infor-
mation is critical for proper counseling and clinical decision
making for cancer survivors considering autotransplantation,
as well as for newly diagnosed oncological patients.
1,5
Here,
we discuss ovarian tissue autotransplantation-related safety
issues and make recommendations for future research and
patient counseling.
Why Is There Concern About the Safety
of Ovarian Tissue Autotransplantation?
The alarm was raised on the safety of ovarian tissue auto-
transplantation in cancer survivors for the first time in 1996.
At that time, healthy mice transplanted with fresh or cryo-
preserved ovarian tissue from mice with lymphoma devel-
oped the disease and died, though one mouse that received
cryopreserved tissue remained healthy.
15
A later xeno-
transplantation study also showed that acute lymphoblastic
leukemia was transmitted to recipient animals via human
ovarian grafts.
16
By using histology or polymerase chain reaction (PCR),
cancer cells have been detected in the ovaries from patients
with leukemia and Ewing sarcoma.
16–18
Apart from these
findings, it is known from clinical experience that different
types of oncological diseases have the potential to metastasize
to the ovaries. For example, breast cancer, a common indica-
tion for ovarian tissue cryopreservation,
9,11–13
has been re-
peatedly demonstrated to metastasize to the ovaries.
19,20
In contrast to these alarming results, other studies report
more reassuring findings when it comes to the safety of
ovarian tissue autotransplantation. Several studies failed to
show the presence of malignant cells in ovarian cortical tissue
Department of Obstetrics and Gynecology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
JOURNAL OF ADOLESCENT AND YOUNG ADULT ONCOLOGY
Volume 2, Number 1, 2013
ªMary Ann Liebert, Inc.
DOI: 10.1089/jayao.2012.0017
31
Journal of Adolescent and Young Adult Oncology 2013.2:31-34.
Downloaded from online.liebertpub.com by 111.185.79.215 on 11/01/15. For personal use only.
from patients with breast cancer,
21,22
lymphoma,
23,24
and
various other oncological diseases.
25
Several authors have tried to provide guidance for clini-
cal decision making and counseling by classifying differ-
ent oncological diseases as having a low, intermediate, or
high risk of ovarian involvement based on the literature
available.
26–28
However, the absolute magnitude of the
risk of ovarian metastasis remains unclear, with some dis-
eases being classified in different risk categories in various
publications.
How to Assess the Risk of Oncological Relapse due
to the Reintroduction of Tumor Cells via Transplantation
One of the pivotal issues when assessing this risk of relapse
is the chance that malignant cells derived from a certain tumor
type and stage are present in the ovaries at the time ovarian
tissue is cryopreserved. Therefore, an overview of epidemio-
logical data on ovarian metastasis in different primary tumor
types, as well as the use of valid diagnostic tools with which
the involvement of the ovaries can be assessed in each indi-
vidual cancer patient, would be useful.
Histology and polymerase chain reaction
Minimal residual disease in ovarian tissue from cancer pa-
tients can bedetected by using histology and/or PCR.
16–18,22–25
During histological examination, the presence of cancer cells
may go unnoticed.
18
PCR is much more sensitive than his-
tology, but tumor-specific PCRs are available for only a lim-
ited number of oncological diseases. In addition, a positive
PCR signal does not provide information about the viability of
tumor cells present in the positive tissue, nor about their
ability to cause relapse after transplantation.
Cortical strips from the same ovary analyzed by PCR may
give different results with respect to the detection of cancer
cells, indicative of sampling bias.
18
As the ovarian strips that
are analyzed by histology or PCR are no longer available for
transplantation purposes, it remains uncertain whether the
strips that are actually used for transplantation purposes are
indeed devoid of cancer cells.
In conclusion, histology and PCR may provide information
about the incidence of minimal residual oncological disease
in the ovarian tissue, but do not guarantee that transplanta-
tion is safe.
Xenotransplantation
Prior to performing an autotransplantation of ovarian
tissue to the human recipient, one or a few cortical fragments
may be xenografted to a suitable immunodeficient host
animal.
15,16,23
Should the recipient animal develop malignant
disease, the tissue obviously contains tumor cells, as with the
mice that developed intraperitoneal leukemic masses after
being grafted with frozen-thawed ovarian tissue from pa-
tients with leukemia.
16
However, if the animals remain heal-
thy, this still offers no guarantee for safe autotransplantation,
as there is again inevitable sample bias.
29
In addition, growth
of the cancer cells might be different in the humans and im-
munocompromised animals.
Follow-up after autotransplantation of ovarian tissue
The duration of follow-up after autotransplantation of
ovarian tissue is still relatively short, and transplantations
have not yet been performed on a large scale.
3,5,8
Therefore,
the safety of autotransplantation cannot be ensured by the
follow-up data currently available. Finally, one should con-
sider that the available follow-up data might be biased by
selective reporting, as no central registration exists.
5
Epidemiological data on the incidence
of ovarian metastases
Follow-up of patients with a certain type of tumor or re-
ports from prophylactic oophorectomy during tumor resec-
tion may provide information about the incidence of ovarian
involvement. However, cancer patient follow-up studies in-
vestigating the incidence of ovarian involvement do not al-
ways clearly describe how they assessed the presence of
ovarian involvement; tumor stage, prior treatment, and other
important prognostic variables may also not be described.
With such a large number of confounding factors left un-
specified, it is quite difficult to extract useful information from
this type of report. Therefore, studies should provide clear
information about tumor types, stages, patient characteristics,
and other relevant prognostic factors, as well as details of the
research methods used. Studies meeting these strict criteria
may be scarce, and a systematic literature search for these
reports is strongly advisable.
One of the difficulties in interpreting epidemiological data
is that these reports include patients who did not have their
ovarian tissue cryopreserved. This means that for a large
number of cases, their ovaries have been exposed to radio- or
chemotherapy that may have eliminated minimal residual
disease in this organ. This is generally not the case in patients
who did opt for ovarian tissue cryopreservation. In addition,
although clinical studies may provide information about the
incidence of clinically relevant ovarian metastases during the
follow-up period, they do not inform about the percentage of
patients who have minimal ovarian involvement at the time
of cancer diagnosis. As a result, it is difficult to estimate the
significance of a single tumor cell in cryopreserved ovarian
tissue, as its capacity to develop into a metastatic lesion after
autotransplantation is unknown.
Conclusion and Recommendations
In conclusion, the available information for counseling
cancer survivors with regard to their risk of oncological re-
lapse after ovarian tissue autotransplantation is insufficient.
In addition, data from different types of studies cannot be
interpreted unambiguously.
It is clear that data on the risk of reintroducing tumor cells
with ovarian transplant are necessary, as a growing number
of patients are expected to request cryopreservation and au-
totransplantation in the near future. We would therefore like
to make the following recommendations.
First, we propose the creation of a database in which all
cancer-related cryopreservation and autotransplantation
cases are registered, including follow-up data. Registration
should preferably take place on an international level. The
European Society of Human Reproduction and Embryology
(ESHRE; Task Force Fertility Preservation), the International
Society for Fertility Preservation (ISFP), or the American So-
ciety for Reproductive Medicine (ASRM) may provide the
necessary infrastructure for such a registry, and would be
logical candidates for setting up this initiative. The
32 BASTINGS ET AL.
Journal of Adolescent and Young Adult Oncology 2013.2:31-34.
Downloaded from online.liebertpub.com by 111.185.79.215 on 11/01/15. For personal use only.
anonymous data from the resulting database should be
available to all clinicians who counsel cancer patients on fer-
tility preservation, ensuring that unbiased follow-up data will
be available as soon as possible.
Next, we suggest performing additional research to im-
prove knowledge and introduce new options into clinical
practice. Studies focusing on the diagnosis and clinical sig-
nificance of minimal residual disease in the ovary will be
pivotal. In addition, further research is needed on techniques
that circumvent the risk of reintroducing cancer cells via au-
totransplantation, such as xenotransplantation of ovarian
tissue followed by IVF, in vitro maturation (IVM),
1,30
and
oocyte formation from stem cells.
31
To date, autotransplanta-
tion of ovarian tissue is the only clinically available option to
restore fertility after ovarian tissue cryopreservation.
1
For
current patient counseling, a systematic review of the available
data on ovarian involvement in neoplastic disease is therefore
urgently needed. Together with the follow-up data from au-
totransplantations, this review will facilitate counseling for
today’s patients who are confronted with fertility preservation
choices.
These recommendations should lead to improvements in
our ability to select the most suitable fertility preservation
option for each individual patient. Until then, we strongly feel
that both clinicians and patients should be aware of the un-
certainties regarding autotransplantation safety.
Acknowledgments
The work of R. Peek was funded by the KiKa Foundation.
Author Disclosure Statement
No competing financial interests exist.
References
1. Jeruss JS, Woodruff TK. Preservation of fertility in patients
with cancer. N Engl J Med. 2009;360(9):902–11.
2. Jadoul P, Dolmans MM, Donnez J. Fertility preservation in
girls during childhood: is it feasible, efficient and safe and to
whom should it be proposed? Hum Reprod Update.
2010;16(6):617–30.
3. Donnez J, Dolmans MM. Preservation of fertility in females
with haematological malignancy. Br J Haematol. 2011;154(2):
175–84.
4. Dittrich R, Lotz L, Keck G, et al. Live birth after ovar-
ian tissue autotransplantation following overnight trans-
portation before cryopreservation. Fertil Steril. 2012;97(2):
387–90.
5. Kolp LA, Hubayter Z. Autotransplantation of cryopreserved
ovarian tissue: a procedure with promise, risks, and a need
for a registry. Fertil Steril. 2011;95(6):1879–86.
6. Akar ME, Carrillo AJ, Jennell JL, Yalcinkaya TM. Robotic-
assisted laparoscopic ovarian tissue transplantation. Fertil
Steril. 2011;95(3):1120.e5–8.
7. Andersen CY, Rosendahl M, Byskov AG, et al. Two
successful pregnancies following autotransplantation of
frozen/thawed ovarian tissue. Hum Reprod. 2008;23(10):
2266–72.
8. Bedaiwy MA, El-Nashar SA, El Saman AM, et al. Re-
productive outcome after transplantation of ovarian tissue: a
systematic review. Hum Reprod. 2008;23(12):2709–17.
9. Lawrenz B, Jauckus J, Kupka MS, et al. Fertility preservation
in >1,000 patients: patient’s characteristics, spectrum, effi-
cacy and risks of applied preservation techniques. Arch
Gynecol Obstet. 2011;283(3):651–6.
10. Meirow D, Baum M, Yaron R, et al. Ovarian tissue cryo-
preservation in hematologic malignancy: ten years’ experi-
ence. Leuk Lymphoma. 2007;48(8):1569–76.
11. Oktay K, Oktem O. Ovarian cryopreservation and trans-
plantation for fertility preservation for medical indications:
report of an ongoing experience. Fertil Steril. 2010;93(3):
762–8.
12. Rosendahl M, Andersen CY, Ernst E, et al. Ovarian function
after removal of an entire ovary for cryopreservation of
pieces of cortex prior to gonadotoxic treatment: a follow-up
study. Hum Reprod. 2008;23(11):2475–83.
13. Sanchez M, Novella-Maestre E, Teruel J, et al. The Valencia
Programme for Fertility Preservation. Clin Transl Oncol.
2008;10(7):433–8.
14. Schmidt KT, Rosendahl M, Ernst E, et al. Autotransplanta-
tion of cryopreserved ovarian tissue in 12 women with
chemotherapy-induced premature ovarian failure: the
Danish experience. Fertil Steril. 2011;95(2):695–701.
15. Shaw JM, Bowles J, Koopman P, et al. Fresh and cryopre-
served ovarian tissue samples from donors with lymphoma
transmit the cancer to graft recipients. Hum Reprod. 1996;
11(8):1668–73.
16. Dolmans MM, Marinescu C, Saussoy P, et al. Reimplanta-
tion of cryopreserved ovarian tissue from patients with
acute lymphoblastic leukemia is potentially unsafe. Blood.
2010;116(16):2908–14.
17. Abir R, Feinmesser M, Yaniv I, et al. Occasional involvement
of the ovary in Ewing sarcoma. Hum Reprod. 2010;25(7):
1708–12.
18. Rosendahl M, Andersen MT, Ralfkiaer E, et al. Evidence of
residual disease in cryopreserved ovarian cortex from fe-
male patients with leukemia. Fertil Steril. 2010;94(6):2186–
90.
19. Bigorie V, Morice P, Duvillard P, et al. Ovarian metastases
from breast cancer: report of 29 cases. Cancer. 2010;116(4):
799–804.
20. Demopoulos RI, Touger L, Dubin N. Secondary ovarian
carcinoma: a clinical and pathological evaluation. Int J Gy-
necol Pathol. 1987;6(2):166–75.
21. Rosendahl M, Timmermans Wielenga V, Nedergaard L,
et al. Cryopreservation of ovarian tissue for fertility preser-
vation: no evidence of malignant cell contamination in
ovarian tissue from patients with breast cancer. Fertil Steril.
2011;95(6):2158–61.
22. Sanchez-Serrano M, Novella-Maestre E, Rosello-Sastre E,
et al. Malignant cells are not found in ovarian cortex from
breast cancer patients undergoing ovarian cortex cryopres-
ervation. Hum Reprod. 2009;24(9):2238–43.
23. Kim SS, Radford J, Harris M, et al. Ovarian tissue har-
vested from lymphoma patients to preserve fertility may
be safe for autotransplantation. Hum Reprod. 2001;16(10):
2056–60.
24. Seshadri T, Gook D, Lade S, et al. Lack of evidence of disease
contamination in ovarian tissue harvested for cryopreser-
vation from patients with Hodgkin lymphoma and analysis
of factors predictive of oocyte yield. Br J Cancer. 2006;94(7):
1007–10.
25. Azem F, Hasson J, Ben-Yosef D, et al. Histologic evaluation
of fresh human ovarian tissue before cryopreservation. Int J
Gynecol Pathol. 2010;29(1):19–23.
SAFETY OF OVARIAN TISSUE AUTOTRANSPLANTATION 33
Journal of Adolescent and Young Adult Oncology 2013.2:31-34.
Downloaded from online.liebertpub.com by 111.185.79.215 on 11/01/15. For personal use only.
26. Akar M, Oktay K. Restoration of ovarian endocrine function
by ovarian transplantation. Trends Endocrinol Metab. 2005;
16(8):374–80.
27. Sonmezer M, Oktay K. Fertility preservation in female pa-
tients. Hum Reprod Update. 2004;10(3):251–66.
28. Sonmezer M, Shamonki MI, Oktay K. Ovarian tissue cryo-
preservation: benefits and risks. Cell Tissue Res. 2005;322(1):
125–32.
29. Bockstaele L, Tsepelidis S, Dechene J, et al. Safety of ovarian
tissue autotransplantation for cancer patients. Obstet Gyne-
col Int. 2012;2012:495142.
30. Oktay K. Ovarian tissue cryopreservation and transplanta-
tion: preliminary findings and implications for cancer pa-
tients. Hum Reprod Update. 2001;7(6):526–34.
31. White YA, Woods DC, Takai Y, et al. Oocyte forma-
tion by mitotically active germ cells purified from ova-
ries of reproductive-age women. Nat Med. 2012;18(3):
413–21.
Address correspondence to:
Lobke Bastings, MD
Department of Obstetrics and Gynecology
Radboud University Nijmegen Medical Centre
PO Box 9101, 6500 HB
Nijmegen
The Netherlands
Email: l.bastings@obgyn.umcn.nl
34 BASTINGS ET AL.
Journal of Adolescent and Young Adult Oncology 2013.2:31-34.
Downloaded from online.liebertpub.com by 111.185.79.215 on 11/01/15. For personal use only.
This article has been cited by:
1. Peek Ron, Westphal Johan R., van Dongen Angelique J.C.M., Loonen Jacqueline J., Braat Didi D.M., Beerendonk Catharina
C.M.. 2014. Fertility Preservation by Ovarian Tissue Cryopreservation After Limited Gonadotoxic Chemotherapy in a 10-Year-
Old Ewing Sarcoma Patient. Journal of Adolescent and Young Adult Oncology 3:2, 92-95. [Abstract] [Full Text HTML] [Full Text
PDF] [Full Text PDF with Links]
2. L. Bastings, C. C. M. Beerendonk, J. R. Westphal, L. F. A. G. Massuger, S. E. J. Kaal, F. E. van Leeuwen, D. D. M. Braat, R.
Peek. 2013. Autotransplantation of cryopreserved ovarian tissue in cancer survivors and the risk of reintroducing malignancy: a
systematic review. Human Reproduction Update . [CrossRef]
Journal of Adolescent and Young Adult Oncology 2013.2:31-34.
Downloaded from online.liebertpub.com by 111.185.79.215 on 11/01/15. For personal use only.
... PARP-1 plays an important role in the cellular response to DNA damage and often regarded as a biomarker of apoptosis. The results Lung is the most common metastasis site of OS. [28] To study the effect of PCP-PEG-ALD nanodrug against pulmonary metastasis of OS, we performed hematoxylin and eosin (H&E) staining [29,30] on the lung of a mouse with orthotopic OS model (Figure 4 E). The experimental outputs showed that amounts of lung-metastatic nodules and proportion of metastatic tumors in the total lung area were reduced in ALD or PEG-ALD treatment groups, especially compared with PBS control groups ( Figure S17). ...
Article
Full-text available
Supramolecular nanodrug assembly driven by supramolecular chemistry is becoming a powerful strategy for medication. The potential of engineered proteins as building blocks for nanoformulations is rarely investigated. Here, we developed a new generation of recombinant protein‐based nanodrug carriers, which is very efficient for loading and delivering the hydrophobic prodrug aldoxorubicin. Significantly enhanced anti‐tumor effects in osteosarcoma (OS) models were observed. The half‐life of the nanodrug reached almost two days and the corresponding bioavailability increased by 17‐fold. This is significantly superior to other drug counterparts, empowering long‐acting OS treatment scenarios. Importantly, off‐target side effects of the prodrug, including cardiotoxicity and lung‐metastasis, were greatly mitigated with our medication. Thus, our assembly strategy enables the customized design of advanced nanodelivery systems employing broader biomaterial building blocks for cancer therapy.
... Several reports provide firm evidence that complete resection of all recurrent osteosarcomas is essential for survival [3,4,[14][15][16][17][18]. The role of other treatments including chemotherapy, immunotherapy and targeted therapies are not fully established yet, but chemotherapy may be able to prolong life in situations where no complete surgical remission can be achieved [3]. ...
Article
The prognosis after relapse of high-grade osteosarcoma is poor and complete resection of all tumors is essential for survival. A 6-year old was diagnosed with high-grade osteosarcoma and treated according to the COSS-96 protocol. Within 5 years from initial diagnosis, five osteosarcoma relapses occurred and every time it was possible to achieve complete surgical remission. Additional treatments included chemotherapy and dendritic cell-based cancer immune therapy. Since the end of therapy of the 5th relapse, he is alive for 11½ years. Our experience further supports that aggressive surgery can help to achieve long-term survival even in patients with multiple osteosarcoma relapses. Pediatr Blood Cancer © 2014 Wiley Periodicals, Inc.
Article
A new generation of protein‐based nanodrugs was developed driven by supramolecular assembly. The nanodrug exhibited significantly enhanced anti‐tumor effects in osteosarcoma models, allowing long‐acting osteosarcoma treatment. Side effects including cardiotoxicity and lung‐metastasis were greatly mitigated with this medication. Abstract Supramolecular nanodrug assembly driven by supramolecular chemistry is becoming a powerful strategy for medication. The potential of engineered proteins as building blocks for nanoformulations is rarely investigated. Here, we developed a new generation of recombinant protein‐based nanodrug carriers, which is very efficient for loading and delivering the hydrophobic prodrug aldoxorubicin. Significantly enhanced anti‐tumor effects in osteosarcoma (OS) models were observed. The half‐life of the nanodrug reached almost two days and the corresponding bioavailability increased by 17‐fold. This is significantly superior to other drug counterparts, empowering long‐acting OS treatment scenarios. Importantly, off‐target side effects of the prodrug, including cardiotoxicity and lung‐metastasis, were greatly mitigated with our medication. Thus, our assembly strategy enables the customized design of advanced nanodelivery systems employing broader biomaterial building blocks for cancer therapy.
Article
Full-text available
Lungs are the primary metastatic sites for osteosarcomas responsible for associated mortality. Recent data has documented role of long non-coding RNAs (lncRNAs) in proliferation and growth of osteosarcoma cells. We evaluated a role of lncRNAs in the lung metastasis of osteosarcoma with the goal of identifying a unique signature. Comparison of different lncRNAs in tumor samples from osteosarcoma with and without lung metastasis led to identification of MALAT1 as the most differentially upregulated lncRNA in the osteosarcoma patients with lung metastasis. MALAT1 was also high in osteosarcoma cells KRIB and MALAT1’s targeted downregulation in these cells led to decreased invasive potential and identification of miR-202 as the miRNA that is sponged by MALAT1. In the lung metastasis in vivo model, parental KRIB cells metastasized to lungs and such metastasis was significantly inhibited in KRIB cells with downregulated MALAT1. Ectopic miR-202 expression attenuated KRIB downregulation-mediated effects on lung metastasis. In yet another in vivo model involving parental SAOS-2 and lung-metastatic derivatives SAOS-2-LM, MALAT1 expression was found to be elevated in lung metastatic cells, which also correlated with reduced miR-202. In conclusion, MALAT1-miR-202 represents a potential lncRNA-miRNA signature that affects lung metastasis of osteosarcomas and could potentially be targeted for therapy.
Article
Full-text available
Girls and young women who require ovariectomy or cancer therapy may consider having their own eggs, embryos or ovarian tissue stored (cryopreserved) for their own future use. Ovarian tissue is simple to collect and contains large numbers of germ cells. Transplantation of fresh and frozen- thawed ovarian tissue in healthy sheep and mice has resulted in normal live young. Similar techniques may be effective in the human but it is unclear whether ovarian tissue cryopreservation and grafting is suitable for ovaries from individuals with cancer or infections. If cancer cells were present in an ovary at the time of collection and survived cryopreservation and grafting they could establish cancer in the recipient. We therefore performed ovarian cryopreservation and transplantation trials using a mouse lymphoma model. This established that the lymphoma was transmitted by grafts of both fresh and frozen ovarian tissue. The normal healthy recipient mice died 9–43 days after receiving a small piece (1 mm of ovarian tissue from a donor with lymphoma. We conclude that ovarian tissue which is collected, cryopreserved and grafted while it contains cancer cells has the potential to spread the cancer to the graft recipient.
Article
Full-text available
Germline stem cells that produce oocytes in vitro and fertilization-competent eggs in vivo have been identified in and isolated from adult mouse ovaries. Here we describe and validate a fluorescence-activated cell sorting-based protocol that can be used with adult mouse ovaries and human ovarian cortical tissue to purify rare mitotically active cells that have a gene expression profile that is consistent with primitive germ cells. Once established in vitro, these cells can be expanded for months and can spontaneously generate 35- to 50-μm oocytes, as determined by morphology, gene expression and haploid (1n) status. Injection of the human germline cells, engineered to stably express GFP, into human ovarian cortical biopsies leads to formation of follicles containing GFP-positive oocytes 1-2 weeks after xenotransplantation into immunodeficient female mice. Thus, ovaries of reproductive-age women, similar to adult mice, possess rare mitotically active germ cells that can be propagated in vitro as well as generate oocytes in vitro and in vivo.
Article
Full-text available
Cancer treatments can induce premature ovarian failure in almost half of young women suffering from invasive neoplasia. Cryopreservation of ovarian cortex and subsequent autotransplantation of frozen-thawed tissue have emerged as promising alternatives to conventional fertility preservation technologies. However, human ovarian tissue is generally harvested before the administration of gonadotoxic treatment and could be contaminated with malignant cells. The safety of autotransplantation of ovarian cortex remains a major concern for fertility preservation units worldwide. This paper discusses the main tools for detecting disseminated cancer cells currently available, their limitations, and clinical relevance.
Article
Full-text available
Ovarian function was studied in Danish patients who had ovarian tissue cryopreserved, and the patients' experiences with the procedure were investigated. There were 92 women who had an entire ovary removed for cryopreservation 18-75 months earlier. Reasons included: breast cancer (n = 31; 34%), Hodgkin's lymphoma (n = 23; 25%), bone marrow transplantation (BMT) (n = 19; 21%) and others (n = 19; 21%). Patients completed a questionnaire, and transvaginal ultrasonic antral follicle count and serum analysis for follicle stimulating hormone (FSH) and anti-Müllerian hormone (AMH) were performed in 73 women. In total, 11% of the BMT patients had normal ovarian function. Hodgkin's patients who only received ABVD (doxorubicin, bleomycin, vinblastine and dacarbazine) (n = 12) and 60% of the breast cancer patients showed little evidence of ovarian damage. Regular menstruation was shown to be a good indicator of ovarian function. The cryopreservation procedure rarely complicated cancer treatment (5%) and 84% felt comforted because they had potentially secured their fertility. Cryopreservation of ovarian tissue should be considered in young female patients with Hodgkin's lymphoma receiving more aggressive treatment than ABVD and in patients scheduled for BMT. The recommendation for breast cancer patient should be individualized. The cryopreservation process did not delay cancer treatment.
Article
Cryopreservation of ovarian tissue for fertility preservation should preferably be performed before the start of anti-cancer therapy. For many patients, however, ovarian tissue preservation is offered after some form of gonadotoxic medication has already been administered. Further, in cases of recurrent malignancies the ovarian tissue may not have been cryopreserved after the initial diagnosis. In this case study, we analyzed the viability of ovarian tissue collected for fertility preservation purposes from a 10-year-old Ewing sarcoma patient after chemotherapy.
Article
BACKGROUND The risk of recurrent oncological disease due to the reintroduction of cancer cells via autotransplantation of cryopreserved ovarian tissue is unknown.METHODSA systematic review of literature derived from MEDLINE, EMBASE and the Cochrane Library was conducted. Studies on follow-up after autotransplantation; detection of cancer cells in ovarian tissue from oncological patients by histology, polymerase chain reaction or xenotransplantation; and epidemiological data on ovarian metastases were included.RESULTSA total of 289 studies were included. Metastases were repeatedly detected in ovarian tissue obtained for cryopreservation purposes from patients with leukaemia, as well as in one patient with Ewing sarcoma. No metastases were detected in ovarian tissue from lymphoma and breast cancer patients who had their ovarian tissue cryopreserved. Clinical studies indicated that one should be concerned about autotransplantation safety in patients with colorectal, gastric and endometrial cancer. For patients with low-stage cervical carcinoma, clinical data were relatively reassuring, but studies focused on the detection of metastases were scarce. Oncological recurrence has been described in one survivor of cervical cancer and one survivor of breast cancer who had their ovarian tissue autotransplanted, although these recurrences may not be related to the transplantation.CONCLUSIONS It is advisable to refrain from ovarian tissue autotransplantation in survivors of leukaemia. With survivors of all other malignancies, current knowledge regarding the safety of autotransplantation should be discussed. The most reassuring data regarding autotransplantation safety were found for lymphoma patients.
Article
To describe the first live birth after transplantation of ovarian tissue following overnight transportation of the tissue before freezing. Technical note. University department of obstetrics and gynecology. A 25-year-old cancer survivor with previous Hodgkin disease and relapse. The ovarian tissue was kept cool for >20 hours in a special transport medium and a special cooling device before it was cryopreserved. After premature ovarian failure due to preconditioning chemotherapy for bone marrow transplantation, the cryopreserved ovarian tissue was transplanted orthotopically. Resumption of ovarian function after transplantation, recovery of fertility, and pregnancy. Ovarian function returned in the patient 3 months after transplantation, as shown by follicle development and estrogen production. During the fifth menstrual cycle, mild stimulation with FSH was initiated in accordance with a low-dose protocol. When ultrasonography revealed a follicle 18-20 mm in size in the ovarian graft, hCG was added and the patient had sexual intercourse at the optimal time point. On day 14 of the luteal phase, hCG concentration and vaginal echography confirmed a viable intrauterine pregnancy, which resulted in a healthy live birth. Overnight transportation of ovarian tissue appears to be possible in combination with appropriate transportation logistics. However, further investigations are needed before this procedure can be offered as a chance for women to preserve fertility independently of direct access to a tissue-processing bank.
Article
Fertility preservation needs have increased dramatically in recent years due to a rising cancer incidence and also a significant increase in survival rates. We report on 200 women participating in the Valencia Programme for Fertility Preservation, of whom 55% were breast cancer patients, 25% Hodgkin's disease patients, and the remaining 20% suffered from other nonmalignant and malignant diseases. Mean age was 28.2 years (11-39). Before patients were submitted to oncological treatment, the right ovarian cortex was extracted by laparoscopy and cryopreserved. Once the patient was free of disease, the right ovarian cortex was thawed and implanted onto the left ovarian medulla (after extraction at the same surgical time of left ovarian cortex). In 95% of cases, a piece approximately 2 x 3 cm was obtained. The procedure did not cause any change in the cancer therapy schedule. Four implants had been performed (all of them in women who underwent chemotherapy prior to ovarian cortex extraction), from which two of the case achieved ovarian function resumption, in one case a month after the implant and in the other 5.5 months after. The remaining two implanted cases were performed 5 and 6 months prior to the writing of this paper, respectively. Ovarian tissue cryopreservation is a feasible option to preserve ovarian function and possibly fertility in adolescents and young women at risk of developing premature ovarian failure (POF) due to chemotherapy and/or radiotherapy.