Method of collecting placental stem cells

Information

  • Patent Grant
  • 9387283
  • Patent Number
    9,387,283
  • Date Filed
    Friday, June 6, 2014
    9 years ago
  • Date Issued
    Tuesday, July 12, 2016
    7 years ago
Abstract
A method of collecting embryonic-like stem cells from a placenta which has been treated to remove residual cord blood by perfusing the drained placenta with an anticoagulant solution to flush out residual cells, collecting the residual cells and perfusion liquid from the drained placenta, and separating the embryonic-like cells from the residual cells and perfusion liquid. Exogenous cells can be propagated in the placental bioreactor and bioactive molecules collected therefrom.
Description
BACKGROUND OF THE INVENTION

1. Field of the Invention


The present invention is generally in the area of stem cell collection, and particularly in the recovery of embryonic-like stem cells and other multipotent stem cells from placentas. These embryonic-like stem cells are derived from the placenta collected after birth. These embryonic-like stem cells have characteristics of embryonic stem cells but are not derived from the embryo.


2. Description of the Background Art


Human stem cells are totipotential or pluripotential precursor cells capable of generating a variety of mature human cell lineages. This ability serves as the basis for the cellular differentiation and specialization necessary for organ and tissue development. Recent success at transplanting such stem cells have provided new clinical tools to reconstitute and/or supplement the bone marrow after myeloablation due to disease, exposure to toxic chemical or radiation. Further evidence exists which demonstrates that stem cells can be employed to repopulate many, if not all, tissues and restore physiologic and anatomic functionality. The application of stem cells in tissue engineering, gene therapy delivery and cell therapeutics is also advancing rapidly.


Obtaining sufficient human stem cells has been problematic for several reasons. First, isolation of normally occurring populations of stem cells in adult tissues has been technically difficult, costly and very limited in quantity. Secondly, procurement of these cells from embryos or fetal tissue including abortuses has raised many ethical and moral concerns. The widely held belief that the human embryo and fetus constitute independent life has justified a moratorium on the use of such sources for any purpose. Alternative sources which do not violate the sanctity of independent life would be essential for further progress in the use of stem cells clinically.


Umbilical cord blood (cord blood) is a known source of hemopoietic pluripotent, progenitor stem cells that are cryopreserved for use in hemopoietic reconstitution. The use of cord blood for this purpose is well known and is becoming a widely used therapeutic procedure. The conventional technique for the collection of cord blood is based on the use of a needle or cannula which is used with the aid of gravity to drain the cord blood from the placenta. Usually the needle or cannula is placed in the umbilical vein and the placenta is gently massaged to aid in draining the cord blood from the placenta. Thereafter the drained placenta has been considered to be of no use and has typically been discarded. A major limitation of stem cell procurement from cord blood has been the frequently inadequate volume of cord blood obtained resulting in insufficient cell numbers to reconstitute bone marrow after transplantation.


Stem cells are in critically short supply. These are important for the treatment of a wide variety of disorders, including malignancies, inborn errors of metabolism, hemoglobinopathies, and immunodeficiences. It would be highly advantageous to have a source of more embryonic stem cells.


Accordingly, it is a primary object of the present invention to provide a method of extracting and recovering hematopoietic stem cells from an exsanguinated placenta.


It is also an object of the invention to provide a method for isolating other embryonic-like and/or omnipotent stem cells from an extractant of a drained placenta.


It is a further object of the invention to provide a method to collect stem cells from the umbilical cord vein, the best source of hemopoietic pluripotent progenitor stem cells.


It is a further object of the present invention to provide a method and means whereby additional embryonic-like stem cells in higher concentrations can be obtained from a drained placenta.


It is a further object of the invention to provide a method of utilizing the isolated and perfused placenta as a biorector providing a good environment for the propagation of endogenous cells, including but not limited to lymphocytes and stem cells.


It is a further object of the present invention to provide a method and means whereby stem cells can be obtained many hours following the birth and expulsion of the placenta from the uterus.


SUMMARY OF THE INVENTION

A method of extracting embryonic-like stem cells from a drained placenta by means of a perfusion technique which utilizes either or both of the umbilical artery and umbilical vein has been developed, on the recovery of human placenta following exsanguination and collection of the residual blood. The placenta is then processed in such a manner as to establish an ex vivo, natural bioreactor environment in which resident stem cells within the parenchyma and extravascular space are recruited and migrate into the empty microcirculation where they can be washed into a collecting vessel by perfusion.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a cross-sectional view of the cannulation of the vein and artery of a placenta to perfuse the placenta and then collect the perfusate.



FIGS. 2a-e are schematics showing the collection, clamping, perfusion, collection and storage of a drained and perfused placenta.



FIG. 3 is a cross-sectional schematic of a perfused placenta in a device for use as a bioreactor.



FIG. 4 is a selection scheme for sorting cells retrieved from a perfused placenta.





DETAILED DESCRIPTION OF THE INVENTION

I. Method of Draining and Extracting Placenta


Draining of Cord Blood and Storage of Fresh Placenta


The method requires access to freshly drained human placentas which have been subjected to a conventional cord blood recovery process by draining substantially all of the cord blood from the placenta. It is important that the placenta be properly stored and drained if it is to be a suitable source of embryonic stem cells. Generally, a placenta should be stored in an anticoagulant solution at a temperature of 5 to 25° C. (centigrade) for no more than 48 hours prior to the collection of the cord blood. Suitable anticoagulant solutions are well known. A preferred anticoagulant solution comprises a solution of heparin (1% w/w in 1:1000 solution). Generally, the drained placenta should be stored for no more than 36 hours before the embryonic-like stem cells are collected.


Extraction of Cells


The preferred embryonic-like stem cell extraction method is based on the perfusion of the drained placenta with a suitable aqueous fluid such as an anticoagulant dissolved in any suitable aqueous isotonic fluid such as 0.9N sodium chloride solution. The anticoagulant for the perfusion liquid may comprise heparin or warfarin sodium at a concentration which is sufficient to prevent the formation of clots of any residual cord blood. Generally from 100 to 1000 units of heparin may be employed.


The extraction procedure is based on the passage of the perfusion liquid through either or both of the umbilical artery and umbilical vein using a gravity flow into the placenta which is suspended in such a manner that the umbilical artery and umbilical vein are at the highest point. It is preferred to connect the umbilical artery and the umbilical vein simultaneously as shown in FIG. 1 to a pipette that is connected via a flexible connector to a reservoir of the perfusion liquid which is passed into the umbilical vein and artery and collected in a suitable open vessel from the surface of the placenta that was attached to the uterus of the mother during gestation.


The collection technique is based on the use of a sufficient amount of the perfusion liquid that will result in the collection of the cells left after the drainage of the cord blood. It has been observed that when the perfusion liquid is first collected, the liquid is colored with the residual red blood cells and tends to become clear as the perfusion liquid is passed through the placenta. Generally from 30 to 100 ml of perfusion liquid is adequate to collect the embryonic-like cells but more or less may be used depending on the observed results.


II. Method of Using the Drained and Perfused Placenta as a BioReactor


Perfusion of the Drained Placenta


As discussed above, the placenta is recovered under aseptic conditions following exsanguination and clamping of the proximal umbilical cord (within 4-5 cm (centimeter) of the insertion into the placental disc) and placental blood recovery and transported in a sterile, thermally insulated (maintaining the temperature of the placenta between 20-28° C.) transport device to the laboratory for processing, for example, by placing the clamped placenta in a sterile zip-lock plastic bag which is then placed in an insulated Styrofoam container or vacuum insulation container, as shown in FIGS. 2a-e.


Use of the Placenta as a Bioreactor


The placenta is placed in a sterile basin and washed with 500 mL of phosphate-buffered normal saline. The wash fluid is discarded. The umbilical vein is cannulated with a Teflon or plastic cannula connected to sterile tubing which is connected to the perfusion manifold, as shown in FIG. 3. The basin is then covered and the placenta is maintained at room temperature (20-25° C.) for a period varying from 2 to 24 hours. The placenta is then perfused at periodically, preferably at 4, 8, 12, and 24 hours, with a volume of perfusate, preferably 100 mL of perfusate (sterile normal saline supplemented with or without 1000 u/L heparin and/or EDTA and/or CPDA (creatine phosphate dextrose)). The effluent fluid which escapes the placenta at the opposite surface is collected and processed to isolate the stem cells of interest. Alterations in the conditions in which the placenta is maintained and the nature of the perfusate can be made to modulate the volume and composition of the effluent.


Stem cells are then isolated from the effluent using techniques known by those skilled in the art, such as for example, density gradient centrifugation, magnet cell separation or other acceptable method, and sorted, for example, according to the scheme shown in FIG. 4.


In variations of this method, the cells in the placenta can be stimulated to produce bioactive molecules, such as immunoglobulin, or other molecules, or stimulated to proliferate, for example, by administration of erythropoietin. The cells can also be genetically engineered prior to harvest, while still in the bioreactor, or at the time of harvest, using for example a viral vector such as an adenoviral or retroviral vector, or using mechanical means such as liposomal or chemical mediated uptake of the DNA.


The procedure is as follows:

  • 1. Fully exsanguinate the placenta and remove any adherent coagulated and non-adherent cellular contaminants.
  • 2. Cultivate and perfuse the placenta with perfusate solution (for example, normal saline solution) with or without an anticoagulant, and/or with or without an antimicrobial agent.
  • 3. Collect both the extravasated perfusate and circulated perfusate into a sterile receptacle.
  • 4. Isolate cell types from the collected perfusate by employing techniques known by those skilled in the art, such as for example, but not limited to density gradient centrifugation, magnet cell separation, affinity cell separation or differential adhesion techniques.


In one embodiment of the invention, the placenta is used as a bioreactor for endogenous cells, including but not limited to lymphocytes and various kinds of pluripotent and/or totipotent stem cells, by incubating the placenta for 48 hours with perfusate solution.


In another embodiment the placenta is processed to remove all endogenous cells and allowing foreign cells to be introduced and propagated in the environment of the perfused placenta. In a particular embodiment the perfused placenta is irradiated with electromagnetic, UV, X-ray, gamma- or beta-radiation to eradicate all remaining viable endogenous cells. The foreign cells of interest to be propagated in the irradiated placental bioreactor are then introduced.


The exogenous cells are induced to propagate by introduction of nutrients and growth factors into the perfusion solution. Serum and other growth factors are added to the propagation perfusion solution or medium. Growth factors are usually proteins and include but are not limited to cytokines, lymphokines, interferons, colony stimulating factors (CSF's), interferons, chemokines, and interleukins. Other growth factors include recombinant human hemopoietic growth factors including ligands, stem cell factors, thrombopoeitin (Tpo), interleukins, and granulocyte colony-stimulating factor (G-CSF). The growth factors introduced into the perfusion solution can stimulate the propagation of undifferentiated stems cells or differentiated hemopoietic cells and stimulate the production of bioactive molecules including but not limited to immunoglobulins, hormones, or other growth factors as previously described.


The present invention will be further understood by reference to the following example.


EXAMPLE 1
Perfusion of Drained Placenta

Twenty ml (milliliter) of phosphate buffered saline solution (PBS) is added to the perfusion liquid and a 10 ml portion is collected and centrifuged for 25 minutes at 3000 rpm (revolutions per minute). The effluent is divided into four tubes and placed in an ice bath. 2.5 ml of a 1% fetal calf serum (FCS) solution in PBS is added and the tubes are centrifuged (140 minutes×10 g (acceleration due to gravity)). The pellet is resuspended in 5 ml of 1% FCS and two tubes are combined. The total mononucleocytes are calculated by adding the total lymphocytes and the total monocytes and multiplying this by the total cell suspension volume.


EXAMPLE 2
Analysis of Cells Obtained by Perfusion and Incubation of Placenta



















WBC

MID

Total
# of



1000/m1
Lym %
%
GRA %
Volume
Cells






















CB (Cord
10.5
43.2
8
48.8
60 ml
6.3 × 108


Blood)


PP (Placenta
12.0
62.9
18.2
18.9
15 ml
1.8 × 108


perfusate,


room


temperature)


PP2
11.7
56.0
19.2
24.8
30 ml
3.5 × 108


(Placenta


perfusate,


37° C.)





Samples of PP were after Ficoll.


Total cell number of PP after Ficoll is 5.3 × 108 and number of CB before processing is 6.3 × 108.


Lym % is percent of lymphocytes;


MID % is percent of midrange white blood cells; and


GRA % is percent of granulocytes.






Cell isolation is achieved by using magnetic cell separation, such as for example, Auto Macs (Miltenyi). Preferably, CD 34+ cell isolation is performed first.


Materials and Methods


Placenta donors were recruited from expectant mothers that enrolled in private umbilical cord blood banking programs and provided informed consent permitting the use of the exsanguinated placenta following recovery of cord blood for research purposes. These donors also permitted use of blinded data generated from the normal processing of their umbilical cord blood specimens for cryopreservation. This allowed comparison between the composition of the collected cord blood and the effluent perfusate recovered using this experimental method described below. All donor data is confidential.


Following exsanguination of the umbilical cord and placenta, the placenta was placed in a sterile, insulated container at room temperature and delivered to the laboratory within 4 hours of birth. Placentas were discarded if, on inspection, they had evidence of physical damage such as fragmentation of the organ or avulsion of umbilical vessels. Placentas were maintained at room temperature (23+/−2° C.) or refrigerated (4° C.) in sterile containers for 2 to 20 hours. Periodically, the placentas were immersed and washed in sterile saline at 25+/−3° C. to remove any visible surface blood or debris. The umbilical cod was transected approximately 5 cm from its insertion into the placenta and the umbilical vessels were cannulated with Teflon or polypropylene catheters connected to a sterile fluid path allowing bi-directional perfusion of the placenta and recovery of the effluent fluid. The system employed in the present invention enabled all aspects of conditioning, perfusion and effluent collection to be performed under controlled ambient atmospheric conditions as well as real-time monitoring of intravascular pressure and flow rates, core and perfusate temperatures and recovered effluent volumes. A range of conditioning protocols were evaluated over a 24 hour post-partum period and the cellular composition of the effluent fluid was analyzed by flow cytometry, light microscopy and colony forming unit assays.


Placental Conditioning


The placenta was maintained under varying conditions in an attempt to simulate and sustain a physiologically compatible environment for the proliferation and recruitment of residual cells. The cannula was flushed with IMDM serum-free medium (GibcoBRL, NY) containing 2 U/ml heparin (EJkins-Sinn, NJ). Perfusion of the placenta continued at a rate of 50 mL per minute until approximately 150 mL of perfusate was collected. This volume of perfusate was labeled “early fraction”. Continued perfusion of the placenta at the same rate resulted in the collection of a second fraction of approximately 150 mL and was labeled “late fraction”. During the course of the procedure, the placenta was gently massaged to aid in the perfusion process and assist in the recovery of cellular material. Effluent fluid was collected from the perfusion circuit by both gravity drainage and aspiration through the arterial cannula.


Placentas were obtained from delivery rooms along with cord blood after obtaining written parental consent, and were processed at room temperature within 12 to 24 hours after delivery. Before processing, the membranes were removed and the maternal site washed clean of residual blood. The umbilical vessels were cumulated with catheters made from 20 gauge Butterfly needles use for blood sample collection. Placentas were then perfused with heparinized (2 U/mL) Dulbecco's modified Eagle Medium (H.DMEM) at the rate of 15 mL/minute for 10 minutes and the perfusates were collected from the maternal sites within one hour and the nucleated cells counted. The perfusion and collection procedures were repeated once or twice until the number of recovered nucleated cells fell below 100/microL. The perfusates were pooled and subjected to light centrifugation to remove platelets, debris and de-nucleated cell membranes. The nucleated cells were then isolated by Ficoll-Hypaque density gradient contrifugation and after washing, resuspended in H.DMEM. For isolation of the adherent cells, aliquots of 5-10×106 cells were placed in each of several T-75 flasks and cultured with commercially available Mesenchymal Stem Cell Growth Medium (MSCGM) obtained from BioWhittaker, and placed in a tissue culture incubator (37° C., 5% CO2). After 10 to 15 days, the non-adherent cells were removed by washing with PBS, which was then replaced by MSCGM. The flasks were examined daily for the presence of various adherent cell types and in particular, for identification and expansion of clusters of fibroblastoid cells.


Cell Recovery and Isolation


Cells are recovered from the perfusates by centrifugation at X 00×g for 15 minutes at room temperature. This procedure served to separate cells from contaminating debris and platelets. The cell pellets were resuspended in IMDM serum-free medium containing 2 U/ml heparin and 2 mM EDTA (GibcoBRL, NY). The total mononuclear cell fraction was isolated using Lymphoprep (Nycomed Pharma, Oslo, Norway) according to the manufacturer's recommended procedure and the mononuclear cell fraction was resuspended. Cells were counted using a hemocytometer. Viability was evaluated by trypan blue exclusion. Isolation of mesenchymal cells was achieved by “differential trypsinization,” using a solution of 0.05% trypsin with 0.2% EDTA (Sigma). Differential trypsinization was possible because fibroblastoid cells detached from plastic surfaces within about five minutes whereas the other adherent populations required more than 20-30 minutes incubation. The detached fibroblastoid cells were harvested following trypsinization and trypsin neutralization, using Trypsin Neutralyzing Solution (TNS, BioWhitaker). The cells were washed in H.DMEM and resuspended in MSCGM. Flow cytometry was carried out using a Becton-Dickinson FACSCalibur instrument and FITC and PE labeled monoclonal antibodies, selected on the basis of known markers for bone marrow-derived MSC (mesenchymal stem cells), were purchased from B.D. and Caltag laboratories (S. San Francisco, Calif.), and SH2, SH3 and Sh4 antibody producing hybridomas were obtained from AM. Cul. and reactivities of the MoAbs in their cultured supernatants were detected by FITC or PE labeled F(ab)′2 goat anti-mouse antibodies. Lineage differentiation was carried out using the commercially available induction and maintenance culture media (BioWhittaker), used as per manufacturer's instructions.


Isolation of Placental Stem Cells


Microscopic examination of the adherent cells in the culture flasks revealed morphologically different cell types. The spindle-shaped cells, the round cells with large nuclei and numerous peri-nuclear small vacuoles and star-shaped cells with several projections, through one of which the cells were attached to the flask. Although no attempts were made to further characterize these adherent cells, similar cells were observed in the culture of bone marrow, cord and peripheral blood, and therefore considered to be non-stem cell in nature. The fibroblastoid cells, appearing last as clusters, were candidates for being MSC and were isolated by differential trypsinization and subcultured in secondary flasks. Phase microscopy of the rounded cells, after trypsinization, to be highly granulated; indistinguishable from the bone marrow-derived MSC produced in the laboratory or purchased from BioWhittaker. When subcultured, the placental-derived cells, in contrast to their earlier phase, adhered within hours, assumed characteristic fibroblastoid shape, and formed a growth pattern identical to the reference bone marrow-derived MSC. Moreover, during subculturing and refeeding, the loosely bound mononuclear cells were washed out and the cultures remained homogeneous and devoid of any visible non-fibroblastoid cell contaminants.


Flow Cytometry


The expression of CD-34, CD-38, and other stem cell-associated surface markers on early and late fraction purified mononuclear cells was assessed by flow cytometry. Briefly, cells were washed in PBS and then double-stained with anti-CD34 phycoerythrin and anti-CD38 fluorescein isothiocyanate (Becton Dickinson, Mountain View, Calif.).


Whereas particular embodiments of this invention have been described herein for purposes of illustration, it will be evident to those persons skilled in the art that numerous variations of the details of the present invention may be made without departing from the invention as defined in the appended claims.

Claims
  • 1. A method of collecting a cell population comprising, perfusing a post-partum human placenta with a perfusion solution by passing said perfusion solution into one or both of the umbilical artery and umbilical vein of said placenta; and collecting said perfusion solution, wherein said collected perfusion solution comprises a cell population comprising a nucleated placental cell population.
  • 2. The method of claim 1, further comprising isolating the cell population comprising a nucleated cell population from the collected perfusion solution.
  • 3. The method of claim 2, wherein the cell population is isolated by centrifugation of the collected perfusion solution.
  • 4. The method of claim 2, further comprising removing platelets from the cell population, thereby producing an isolated nucleated placental cell population.
  • 5. The method of claim 4, wherein the nucleated cell population is isolated by density gradient centrifugation.
  • 6. The method of claim 4, wherein the collected perfusion solution consists of perfusion solution that has been collected from umbilical vessels.
  • 7. The method of claim 4, wherein the collected perfusion solution comprises perfusion solution that has been collected from umbilical vessels and perfusion solution that has been collected from maternal sites of the placenta.
  • 8. The method of claim 1, wherein the perfusion solution comprises heparin.
  • 9. The method of claim 1, wherein collecting the perfusion solution comprises collection via gravity drainage.
  • 10. The method of claim 1, wherein collecting the perfusion solution comprises collection via aspiration.
  • 11. The method of claim 1, wherein neither said cell population nor said nucleated cell population are combined with cord blood or hematopoietic stem cells from cord blood.
BENEFIT OF PRIOR PROVISIONAL APPLICATION

This application is a divisional of U.S. application Ser. No. 12/618,664, filed Nov. 13, 2009, which is a continuation of U.S. application Ser. No. 11/187,400 (now U.S. Pat. No. 7,638,141), filed Jul. 21, 2005, which is a continuation of U.S. application Ser. No. 10/004,942 (now U.S. Pat. No. 7,045,148), filed Dec. 5, 2001, which claims benefit of U.S. Provisional Application No. 60/251,900, filed Dec. 6, 2000, the specifications of which are each incorporated herein by reference.

US Referenced Citations (140)
Number Name Date Kind
3862002 Sanders Jan 1975 A
4829000 Kleinman et al. May 1989 A
5004681 Boyse et al. Apr 1991 A
5192553 Boyse et al. Mar 1993 A
5372581 Anderson Dec 1994 A
5415665 Hessel et al. May 1995 A
5635387 Fei et al. Jun 1997 A
5665557 Murray et al. Sep 1997 A
5670351 Emerson et al. Sep 1997 A
5827735 Young et al. Oct 1998 A
5855620 Bishopric et al. Jan 1999 A
5866414 Badylak et al. Feb 1999 A
5874301 Keller et al. Feb 1999 A
5879318 Van Der Heiden et al. Mar 1999 A
5905041 Beug et al. May 1999 A
5914108 Tsukamoto et al. Jun 1999 A
5914268 Keller et al. Jun 1999 A
5916202 Haswell Jun 1999 A
5919176 Kuypers et al. Jul 1999 A
5928214 Rubinstein et al. Jul 1999 A
5993429 Kuypers et al. Nov 1999 A
6030836 Thiede Feb 2000 A
6059968 Wolf, Jr. May 2000 A
6110739 Keller et al. Aug 2000 A
6179819 Haswel Jan 2001 B1
6190368 Kuypers et al. Feb 2001 B1
6255112 Thiede et al. Jul 2001 B1
6326019 Tseng Dec 2001 B1
6461645 Boyse et al. Oct 2002 B1
6800480 Bodnar et al. Oct 2004 B1
7045148 Hariri May 2006 B2
7147626 Goodman Dec 2006 B2
7244759 Muller et al. Jul 2007 B2
7255879 Hariri Aug 2007 B2
7311904 Hariri Dec 2007 B2
7311905 Hariri Dec 2007 B2
7468276 Hariri Dec 2008 B2
7498171 Hariri et al. Mar 2009 B2
7638141 Hariri Dec 2009 B2
7682803 Paludan et al. Mar 2010 B2
7700090 Heidaran et al. Apr 2010 B2
7909806 Goodman Mar 2011 B2
7914779 Hariri Mar 2011 B2
7928280 Hariri et al. Apr 2011 B2
7976836 Hariri Jul 2011 B2
7993918 Paludan et al. Aug 2011 B2
8057788 Hariri Nov 2011 B2
8057789 Hariri Nov 2011 B2
8071135 Liu et al. Dec 2011 B2
8071376 Heidaran Dec 2011 B2
8105634 Liu et al. Jan 2012 B2
8202703 Edinger et al. Jun 2012 B2
8263065 Zhang et al. Sep 2012 B2
8986984 Hariri Mar 2015 B2
20020123141 Hariri Sep 2002 A1
20020132343 Lum Sep 2002 A1
20020160510 Hariri Oct 2002 A1
20030032179 Hariri Feb 2003 A1
20030180269 Hariri Sep 2003 A1
20030187515 Hariri Oct 2003 A1
20030235909 Hariri Dec 2003 A1
20040028660 Hariri et al. Feb 2004 A1
20040048372 Hariri Mar 2004 A1
20040048796 Hariri et al. Mar 2004 A1
20040171147 Hariri Sep 2004 A1
20040219136 Hariri Nov 2004 A1
20050019908 Hariri Jan 2005 A1
20050118715 Hariri et al. Jun 2005 A1
20050143420 Moutouh-de Parseval et al. Jun 2005 A1
20050148034 Hariri et al. Jul 2005 A1
20050266391 Bennett et al. Dec 2005 A1
20050272148 Hariri Dec 2005 A1
20050276792 Kaminski et al. Dec 2005 A1
20060060494 Goodman et al. Mar 2006 A1
20070020225 Abramson et al. Jan 2007 A1
20070021704 Hariri et al. Jan 2007 A1
20070021762 Liu et al. Jan 2007 A1
20070038298 Sulner et al. Feb 2007 A1
20070043328 Goodman et al. Feb 2007 A1
20070053888 Hariri Mar 2007 A1
20070092497 Hariri Apr 2007 A1
20070134210 Heidaran Jun 2007 A1
20070190034 Paludan et al. Aug 2007 A1
20070190042 Edinger et al. Aug 2007 A1
20070275362 Edinger et al. Nov 2007 A1
20070292399 Heidaran et al. Dec 2007 A1
20070292910 Heidaran et al. Dec 2007 A1
20080032401 Edinger et al. Feb 2008 A1
20080044848 Heidaran Feb 2008 A1
20080069895 Liu et al. Mar 2008 A1
20080131410 Hariri Jun 2008 A1
20080131522 Liu et al. Jun 2008 A1
20080131966 Hariri Jun 2008 A1
20080152624 Paludan et al. Jun 2008 A1
20080152629 Edinger et al. Jun 2008 A1
20080175824 Heidaran et al. Jul 2008 A1
20080181935 Bhatia et al. Jul 2008 A1
20080181967 Liu et al. Jul 2008 A1
20080206343 Edinger et al. Aug 2008 A1
20080208158 Goodman et al. Aug 2008 A1
20080213228 Edinger et al. Sep 2008 A1
20080226595 Edinger et al. Sep 2008 A1
20090053805 Hariri Feb 2009 A1
20090104164 Zhang et al. Apr 2009 A1
20090136471 Heidaran et al. May 2009 A1
20090142831 Hariri Jun 2009 A1
20090226406 Hariri Sep 2009 A1
20090252710 Zhang et al. Oct 2009 A1
20100047213 Zeitlin et al. Feb 2010 A1
20100047214 Abramson et al. Feb 2010 A1
20100047351 Zeitlin et al. Feb 2010 A1
20100120015 Hariri May 2010 A1
20100124569 Abbot May 2010 A1
20100143312 Hariri Jun 2010 A1
20100172830 Heidaran Jul 2010 A1
20100183571 Paludan et al. Jul 2010 A1
20100260847 Hariri Oct 2010 A1
20100291679 Edinger et al. Nov 2010 A1
20100297689 Edinger et al. Nov 2010 A1
20100323446 Barnett et al. Dec 2010 A1
20110003387 Abbot et al. Jan 2011 A1
20110206645 Zhang et al. Aug 2011 A1
20110217271 Hariri Sep 2011 A1
20110217272 Hariri Sep 2011 A1
20110223141 Hariri Sep 2011 A1
20110250182 Abbot Oct 2011 A1
20110250185 Paludan et al. Oct 2011 A1
20110280843 Edinger et al. Nov 2011 A1
20110280845 Edinger et al. Nov 2011 A1
20110280849 Zhang et al. Nov 2011 A1
20110311491 Edinger et al. Dec 2011 A1
20110318401 Hariri et al. Dec 2011 A1
20120020936 Hariri Jan 2012 A1
20120034195 Hariri Feb 2012 A1
20120058089 Hariri Mar 2012 A1
20120121550 Heidaran May 2012 A1
20120148553 Hariri et al. Jun 2012 A1
20120171161 Abramson et al. Jul 2012 A1
20120171295 Abramson et al. Jul 2012 A1
20120230959 Abbot et al. Sep 2012 A1
Foreign Referenced Citations (3)
Number Date Country
WO 0017325 Mar 2000 WO
WO 0073421 Dec 2000 WO
WO 0121767 Mar 2001 WO
Non-Patent Literature Citations (35)
Entry
Wang et al. 2010, Morgan & Claypool Life Sciences; Chapter 2, Placental Blood Circulation.
U.S. Appl. No. 13/473,509, filed May 16, 2012, Edinger et al.
U.S. Appl. No. 13/480,370, filed May 24, 2012, Edinger et al.
U.S. Appl. No. 13/485,161, filed May 31, 2012, Herzberg et al.
U.S. Appl. No. 13/584,612, filed Aug. 13, 2012, Hariri, et al.
Addison, et al., 1991, “Metabolism of Prednisolone by the Isolated Perfused Human Placental Lobule,” J. Ster. Biochem. Mol. Biol., 39(1):83-90.
Belvedere, et al., 2000, “Increased Blood Volume and CD34(+)CD38(−) Progenitor Cell Recovery Using a Novel Umbilical Cord Blood Collection System,” Stem Cells 18(4):245-251.
Bersinger, et al., 1992, “Effect of Late Pregnancy Serum on the Synthesis and Release of Pregnancy Proteins by the Perfused Human Term Placenta,” Reprod. Fertil. Dev. 4:585-588.
Contractor, et al., 1984, “A comparison of the effects of different perfusion regimens on the structure of the isolated human placental lobule,” Cell Tissue Res. 237:609-617.
Heidaran, Disclosure Document No. 457045 for “A Method or Process for the Treatment of Degenerative Conditions or Cancer Employing Custom Fabricated Organ Tissue Grafts Using Cells Isolated, Expanded, and Stored at Birth”, 15 pages, stamped received by OIPE on May 28, 1999, paper dated May 13, 1999.
Jaroscak et al., 2000, “Preliminary characterization of the surface staining of placental derived adherent cells: a potential new source of stroma for umbilical cord blood (UCB) expansion,” Blood 96(11, Pt 2).
Ma et al., 1999, “Development of an in vitro Human Placenta Model by the Cultivation of Human Trophoblasts in a Fiber-Based Bioreactor System,” Tissue Engineering 5, 91-102.
Minguell, et al., 2001, “Mesenchymal Stem Cells,” Exp. Biol. Med. 226:507-520.
Muhlemann, et al., 1995, “Cytomegalovirus in the perfused human term placenta in vitro,” Placenta 16:367-373.
Nagayama et al., 1999, “Immunological reconstitution after cord blood transplantation for an adult patient”, Bone Marrow Transplantation 24: 211-213.
Papaioannou, et al., 2004, “Stem Cells from Early Mammalian Embryos” Stem Cells Handbook:19-31.
Webster, 2004, Tissue Culture: Merriam-Webster's Online Dictionary.
Wikipedia, Placenta, 2014.
Gangal et al., 2010, “Organ culture”, In Principles and Practice of animal tissue culture, 2nd edition by Sudha Gangal, p. 51, Chapter 9.
Armant et al., 1994, J Exp Zoology, 269:146-156.
Devlin et al., 1985, “In vitro metabolism of 25-hydroxycholecalciferol by isolated cells from human deciduas”, J Clin Endocrinol Metab, 60(3):880-885.
Erices et al., 2000, “Mesenchymal progenitor cells in human umbilical cord blood”, Brit J Haematol, 109:235-242.
Dorrell, 2000, “Expansion of human cord blood CD34+CD38− cells in ex vivo culture during retroviral transduction without a corresponding increase in SCID repopulating cell (SRC) frequency: dissociation of SRC phenotype and function”, Blood, 95(1):102-110.
Emerson, 1996, “Ex vivo expansion of hematopoietic precursors, progenitors, and stem cells: the next generation of cellular therapeutics”, Blood, 87(8):3082-3088.
Srour et al., 1999, “Ex vivo expansion of hematopoietic stem and progenitor cells: are we there yet?”, The Journal of Hematotherapy, 8:93-102.
Hatzopoulos et al., 1998, “Isolation and characterization of endothelial progenitor cells from mouse embryos”, Development, 125:1457-1468.
Shamblott et al., 1998, “Derivation of pluripotent stem cells from cultured human primordial germ cells”, Proc Natl Acad Sci USA, 95:13726-13731, Erratum in: Proc Natl Acad Sci USA, 96:1162(1999).
Thompson et al., 1998, “Embryonic stem cell lines derived from human blastocysts”, Science, 282:1145-1147. Erratum in: Science, 282:1827.
Turner et al., 1992, “A modified harvest technique for cord blood hematopoietic stem cells”, Bone Marrow Transplant, 10:89-91.
Wang et al., 2001, “Enhanced recovery of hematopoietic progenitor and stem cells from cultivated postpartum human placenta”, Blood, 98(11/1):183a, abstract No. 769.
Ye et al., 2001, “Recovery of placental-derived adherent cells with mesenchymal stem cell characteristics”, Blood, 98(11/1):147b, abstract No. 4260.
Myllynen, 2003, “In search of models for hepatic and placental pharmacokinetics”, dissertation, University of Oulu.
Van Bekkum, 1990, “The pluripotent hemopoietic stem cell: its identification and applications”, Verh Dtsch Ges Pathol, 74:19-24.
Caplan, 1994, “The mesengenic process”, Clin Plast Surg, 21(3):429-435.
Moore, 1997, “A simple perfusion technique for isolation of maternal intervillous blood mononuclear cells from human placentae,” J. Immunol. Methods 209:93-104.
Related Publications (1)
Number Date Country
20140349391 A1 Nov 2014 US
Provisional Applications (1)
Number Date Country
60251900 Dec 2000 US
Divisions (1)
Number Date Country
Parent 12618664 Nov 2009 US
Child 14298294 US
Continuations (2)
Number Date Country
Parent 11187400 Jul 2005 US
Child 12618664 US
Parent 10004942 Dec 2001 US
Child 11187400 US