干细胞之家 - 中国干细胞行业门户第一站

 

 

搜索
朗日生物

免疫细胞治疗专区

欢迎关注干细胞微信公众号

  
查看: 365523|回复: 236
go

Placental Growth Factor-1 Potentiates Hematopoietic Progenitor Cell Mobilization [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:59 |只看该作者 |倒序浏览 |打印
作者:Carmelo Carlo-Stellaa,b, Massimo Di Nicolaa, Paolo Longonia, Loredana Clerisc, Cristiana Lavazzaa, Raffaella Milanid, Marco Milanesia, Michele Magnia, Virgilio Pacee, Francesco Colottae, Maria A. Avanzinif, Franca Formellic, Alessandro M. Giannia,b作者单位:aCristina Gandini Medical Oncology Unit, Istituto Nazionale Tumori, Milano, Italy;bChair of Medical Oncology, University of Milano, Milano, Italy;cDepartment of Experimental Oncology, Istituto Nazionale Tumori, Milano, Italy;dHematology and Bone Marrow Transplantation Unit, Istituto Nazionale Tumori
' V( e$ ^$ q3 H0 ?                  
4 j7 G9 I# p/ h# w                  
- W$ c4 u0 }; s8 H. Y. ?         
! X4 J6 J7 i% T# v: I                         ' @1 G% W6 L/ q! P, |
            
( M* k; S1 @" r' C7 j$ B            + E& X8 \2 \' W4 U- Z
            3 ~4 i8 E5 q" F9 C
            , ~( @1 z3 e; k
                      7 b! d9 i6 Y% k3 ~. _
        $ V- E: ?( p9 u* \  Y# ~
        , w# J: E* o6 a
        & A# J9 y/ _( x: A
          【摘要】
/ N4 N! Y' D! A. ~      The complex hematopoietic effects of placental growth factor (PlGF) prompted us to test in mice and nonhuman primates the mobilization of peripheral blood progenitor cells (PBPCs) elicited by recombinant mouse PlGF-2 (rmPlGF-2) and recombinant human PlGF-1 (rhPlGF-1). PBPC mobilization was evaluated by assaying colony-forming cells (CFCs), high-proliferative potential-CFCs (HPP-CFCs), and long-term culture-initiating cells (LTC-ICs). In mice, both rmPlGF-2 and rhPlGF-1 used as single agents failed to mobilize PBPCs, whereas the combination of rhPlGF-1 and granulocyte colony-stimulating factor (rhG-CSF) increased CFCs and LTC-ICs per milliliter of blood by four- and eightfold, respectively, as compared with rhG-CSF alone. rhPlGF-1 plus rhG-CSF significantly increased matrix metalloproteinase-9 plasma levels over rhG-CSF alone, suggesting a mechanistic explanation for rhPlGF-1/rhG-CSF synergism. In rhesus monkeys, rhPlGF-1 alone had no mobilization effect, whereas rhPlGF-1 (260 µg/kg per day) plus rhG-CSF (100 µg/kg per day) increased rhG-CSF-elicited mobilization of CFCs, HPP-CFCs, and LTC-ICs per milliliter of blood by 5-, 7-, and 15-fold, respectively. No specific toxicity was associated with the administration of rhPlGF-1 alone or in combination. In conclusion, our data demonstrate that rhPlGF-1 significantly increases rhG-CSF-elicited hematopoietic mobilization and provide a preclinical rationale for evaluating rhPlGF-1 in the clinical setting.
6 a3 N8 |5 |5 W2 n3 n3 B          【关键词】 Stem cell mobilization Stem cell transplantation Placental growth factor Granulocyte colony-stimulating factor2 }5 b/ I$ Y4 `# J- x( E
                  INTRODUCTION( _! L7 f5 a$ u3 \1 c% c2 E
& |8 r7 u% e+ b4 d" U
Mobilized peripheral blood progenitor cells (PBPCs) have an established role in the management of patients with a variety of cancers undergoing autologous .: O% ?/ M. e$ W; |+ b" b

0 g2 l% y9 @  e2 lAlthough suboptimal mobilization represents a substantial limitation to the completion of cell therapy programs requiring high amounts of CD34  cells (i.e., haploidentical bone marrow transplanation, tandem autografts, gene therapy on hematopoietic stem cells), the lack of autologous stem cells raises important issues for the clinical management of patients for whom autologous SCT has proven to be clinically beneficial. Therefore, any procedure applicable to cancer patients and capable of increasing the yield of circulating progenitors in the absence of added toxicity, is expected to have a profound impact on the feasibility, toxicity, and costs of hematopoietic transplants. Despite several attempts to improve PBPC mobilization by molecules capable of interfering with the mechanism(s) regulating hematopoietic stem cell trafficking . Exploitation of the full clinical potential of these new agents is still under evaluation, and no general consensus exists for a therapy capable of improving stem cell mobilization.
8 {3 ?! g4 s3 b! T- ]8 K
! e% ^- M% a3 }Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family, occurring in at least four splicing isoforms, PlGF-1 to PlGF-4, which differ in size and binding properties .
& ?1 O. @4 E' z- N9 W2 Q1 p. d7 Q" z1 _# h0 _8 A# m) [- c! \
These observations suggesting that PlGF might exert a relevant clinical impact on SCT by improving stem cell mobilization prompted us to test the capability of PlGF to mobilize PBPCs in BALB/c mice and in a nonhuman primate model allowing the simulation of PBPC mobilization as occurs in a clinical situation. Hematopoietic mobilization was analyzed by assaying committed colony-forming cells (CFCs), high-proliferative potential progenitors (HPP-CFCs), as well as long-term culture-initiating cells (LTC-ICs) in mice and nonhuman primates treated with PlGF alone or in combination with rhG-CSF.
' T5 w) T% I- [" R( G6 U& e1 M. ?5 B. P0 {( d% ~
MATERIALS AND METHODS+ Q6 I2 r& K; a
- ~# S  \1 T# _" k' A3 h% J  W
Studies in Mice9 K6 R, `2 m  k+ F5 S& j

4 K. ^/ E( p* i: s/ \; m4 DAnimals.   Six- to 8-week-old female BALB/c mice, with a body weight of 20¨C25 g, were purchased from Charles River Laboratories (Milan, Italy, http://www.criver.com). Experimental procedures performed on animals were approved by the Ethical Committee for Animal Experimentation of the Istituto Nazionale Tumori (Milan, Italy) and were carried out in accordance with the guidelines of the United Kingdom Coordinating Committee on Cancer Research .
$ o% Y/ \- h# ^. W7 ?$ p
7 m  v" ~  z/ M% B/ Y' e2 B7 i# cCytokines.   rhG-CSF (Neupogen; Amgen, Thousand Oaks, CA, http://www.amgen.com) was from Roche S.p.A. (Milan, Italy, http://www.roche.it), recombinant mouse PlGF-2 (rmPlGF-2) was purchased from R&D Systems Europe Ltd. (Abingdon, U.K., http://www.rndsystems.com) and recombinant human PlGF-1 (rhPlGF-1) was a kind gift of Dr. A. Mion (Geymonat S.p.A., Anagni, Italy, http://www.geymonat.com).9 o3 |9 a* D4 C- G4 W0 @% p( o

9 p/ S" z9 o/ K* s- Z" ?1 rMobilization Protocols.   The standard mobilization protocol known to elicit a maximal PBPC mobilization consisted of i.p. injection of 10 µg/day rhG-CSF (days 1¨C5) . Single-agent treatments with PlGF consisted of i.p. injection of 5 µg/day rmPlGF-2 (days 1¨C5) or 10 µg/day rhPlGF-1 (days 1¨C5). Combination treatments consisted of 5-day treatments with rhG-CSF (10 µg/day) plus either rmPlGF-2 (2.5, 5, or 10 µg/day) or rhPlGF-1 (5, 10, or 15 µg/day). The dose ranges of PlGF used in combination studies had been identified in preliminary experiments as the doses inducing a maximal enhancement of rhG-CSF-elicited PBPC mobilization. Control mice were injected with low-endotoxin phosphate-buffered saline (PBS) containing 0.1% mouse serum albumin (MSA) as a carrier. Each experiment was performed on at least three separate occasions, and four mice per group and time point were used. Unless otherwise stated, all animal groups were sacrificed 2¨C3 hours after the last treatment.
3 A! k' P8 j' y/ b& C  S3 N8 X* L& k' q  \
Cell Harvesting and Separation.   Peripheral blood was harvested from the orbital plexus into heparin-containing tubes. After white blood cell (WBC) counting, blood was diluted (1:4, vol/vol) with PBS, and mononuclear cells (MNCs) were separated by centrifugation (280g, 30 minutes, room temperature) on a Ficoll discontinuous density gradient . Cells were then washed twice in Iscove's modified Dulbecco's medium (Cambrex Bio Science Verviers, S.p.r.l., Verviers, Belgium, http://www.cambrex.com) supplemented with 10% fetal bovine serum (FBS) (StemCell Technologies, Vancouver, BC, Canada, http://www.stemcell.com), 2 mM L-glutamine, and antibiotics.6 }0 F! i9 l5 g0 ^) T

9 ~7 |+ v4 u& a3 O# Y- SColony-Forming Cell Assay.   Total colony-forming cells (CFCs)¡ªgranulocyte-macrophage colony-forming units (CFU-GM), erythroid burst-forming units (BFU-E), and multilineage CFU (CFU-GEMM)¡ªwere assessed in methylcellulose cultures .
6 }0 y7 |$ _. m+ h( V, o( A; P; J: ]! I+ L6 X
LTC-IC Assay.   LTC-ICs were assessed in bulk cultures ., w5 ~  i  Q) Z; k- O
0 H" O* D$ C, M) h& k! o& o: O6 U
Assay for Radioprotective Cells.   Radioprotective cells were studied by comparing the survival of lethally irradiated (850 cGy) BALB/c mice transplanted with MNCs obtained from syngeneic donors that had been treated for 5 days with PBS/MSA, rhG-CSF (10 µg/day), or rhPlGF-1 (10 µg/day) plus rhG-CSF (10 µg/day). Ten recipient mice were injected with control MNCs from PBS/MSA-treated animals, and 15 mice were injected with 2 x 105 or 5 x 105 rhG-CSF- or rhPlGF-1/rhG-CSF-mobilized cells. Control irradiated mice were included in all experiments. A dose of irradiation (850 cGy) that in preliminary experiments resulted in 100% mortality by day 15 was chosen. Recipient mice were placed in a polymethylmetaacetate (PMMA) box and given total body irradiation by a Radgil x-ray treatment unit (Gilardoni, Milan, Italy, http://www.gilardoni.it), supplied with a 200-kV x-ray unit, at a dose rate of 35 cGy/minute. Recipient mice were injected via the tail vein 2¨C3 hours after irradiation .& t0 E, L  i0 U" _

+ d5 Z# ]* u  xImmunoassays of Cytokines.   Plasma levels of human PlGF and mouse promatrix metalloproteinase-9 (pro-MMP-9) were evaluated using commercial enzyme-linked immunosorbent assay (R&D Systems Europe Ltd.) in accordance with the manufacturer's instructions." J2 S' M0 V# c+ F2 v! F) ], A

3 q+ C1 I8 M; P/ _5 WStudies in Nonhuman Primates4 Z3 C0 K" W/ c/ N' Y

+ J4 q! }$ W7 d$ \1 o9 _: T) ~9 c1 ~Animals.   Male rhesus monkeys (Macaca mulatta) ranging in age from 4 to 6 years and with a mean weight of 5 ¡À 0.6 kg were housed in individual stainless steel cages in conventional holding rooms at the Biomedical Primate Research Centre (BPRC) (Rijswijk, The Netherlands, http://www.bprc.nl). Animals had no experimental history of previous antibody or cytokine administration. Monkeys were housed in accordance with guidelines of the Animal Care and Use Committee of the BPRC and were provided with commercial monkey chow (Hope Farms, Woerden, The Netherlands, http://www.hopefarms.nl) supplemented with fresh fruit, vegetables, and bread. Tap water was available ad libitum via an automatic watering system. The study was approved by the Animal Care and Use Committee of the BPRC.3 @) I1 H, |9 i& T

0 h% \/ `8 x, y" F( J. V0 eMobilization Protocols.   A cohort of rhesus monkeys (n = 4) received four consecutive mobilization cycles separated from one another by a 6-week washout period. At cycle 1, animals received a standard mobilization protocol consisting of subcutaneous injection of rhG-CSF (100 µg/kg per day for 5 days). This regimen is known to elicit a maximal PBPC mobilization .
" }% j7 C, ~4 S1 S- c1 G/ Z" U/ ]
/ e3 k/ s2 A5 y2 x1 F9 YEvaluation of Rhesus Monkey Blood.   Mobilization was evaluated by complete blood counts (CBCs), differential counts, and frequency and absolute numbers of CFCs, HPP-CFCs, and LTC-ICs. Mobilization parameters were analyzed daily during treatment (days 1¨C5) and 3 and 5 days after cessation of therapy. Peripheral blood samples were obtained from the femoral vein of anesthetized primates (ketamin, 10 mg/kg, intramuscularly) using aseptic techniques.
' [2 e& A. A1 d: W. ?; X2 r$ ?! R; J. e+ X4 B
CBCs and Differential Counts.   CBCs were performed using EDTA-anticoagulated blood and an automated counter (Sysmex SF-3000; Goffin Meyvis, Ettenleur, The Netherlands, http://www.goffinmeyvis.com). Differential counts were performed on Wright-Giemsa-stained blood smears.
1 R0 G; W5 o8 w3 ?0 n
, \' h: R0 {6 p% g; |9 jCFC and HPP-CFC Assays.   Total CFCs (i.e., CFU-GM, BFU-E, plus CFU-GEMM) and HPP-CFCs were assayed according to a previously described technique . The absolute number of circulating CFCs or HPP-CFCs in blood is a function of the frequency of CFCs or HPP-CFCs multiplied by the total number of MNCs per milliliter of blood.: H0 D& u( P; S" e

$ D% X5 t5 U' U+ tLTC-IC Assays.   The frequency of LTC-ICs was assessed under limiting dilution conditions ., P  x  d; c# c/ ?3 s8 i
& T7 D5 G' g5 f& ?+ q' _
Statistical Analysis.   Four plates were scored for each data point per experiment, and the results were expressed as the mean ¡À 1 SEM. The Student's t test for unpaired or paired data (two-tail) was used as appropriate to test the probability of significant differences among different mobilization cycles. Differences were considered significant if p  .05. Statistical analysis was performed with the statistical package Prism 4 (GraphPad Software, Inc., San Diego, http://www.graphpad.com) run on a Macintosh G4 personal computer (Apple Computer, Inc., Cupertino, CA, http://www.apple.com). The LTC-IC frequencies were calculated from the proportion of wells that were negative using L-Calc software, which uses Poisson statistics and the method of maximum likelihood.
6 W9 J7 I; ~+ J6 u- v! f& k& \9 K, x- F7 K0 L; }) }* n
RESULTS
1 g- N' b) D" B8 ^$ G% Z: L6 ?) [0 u* _) z
Effect of Murine PlGF-2 on rhG-CSF-Elicited PBPC Mobilization in BALB/c Mice# Q: R5 X* c1 Z1 M+ C

9 h  T; a4 O& ~+ `0 YIn contrast to previous studies demonstrating that administration of an adenoviral vector expressing PlGF induced a significant PBPC mobilization , a 5-day i.p. treatment of BALB/c mice with rmPlGF-2 alone had no effect on hematopoietic mobilization (Table 1). However, the complex hematopoietic and microenvironmental effects of PlGF strongly suggesting that this cytokine may affect stem cell mobilization, prompted us to investigate whether rmPlGF-2 could eventually enhance rhG-CSF-elicited PBPC mobilization., b: q/ s8 ]' D5 D0 V5 _/ y% K; k

+ Q7 T) g$ i! P! S! {, uTable 1. Hematopoietic mobilization in BALB/c mice upon treatment with rhG-CSF and rmPlGF-27 K! t$ A& s+ k) }9 u4 O

% N9 @+ q/ G4 K( U- G2 NA 5-day combination therapy with rmPlGF-2 (2.5 and 5 µg/day) and rhG-CSF (10 µg/day) had no effect on WBC counts but enhanced in a dose-dependent manner rhG-CSF-elicited PBPC mobilization (Table 1). As compared with rhG-CSF alone, a 5-day course of 5 µg/day rmPlGF-2 plus 10 µg/day rhG-CSF increased CFC frequency and absolute numbers and LTC-IC absolute numbers by twofold (p  .0002), twofold (p  .01), and sevenfold (p  .0002), respectively. Using this combination regimen (i.e., 10 µg/day of rhG-CSF plus 5 µg/day of rmPlGF-2), the median absolute numbers of CFCs and LTC-ICs were approximately 6,000 and 1,200 per milliliter, respectively. Increasing the dose of rmPlGF-2¨C10 µg/day failed to further improve rhG-CSF-elicited PBPC mobilization (data not shown).! B+ r. j! w+ w" O' ]1 x9 c7 [$ }
: y7 G2 t# f3 p$ A- t  D7 Z
Effect of Human PlGF-1 on rhG-CSF-Elicited PBPC Mobilization in BALB/c Mice
6 o; O) x: o  o% e) Z, {& d6 u
2 |% m) N0 m( o8 ASimilarly to what was observed for rmPlGF-2, treatment of BALB/c mice with rhPlGF-1 alone had no mobilizing activity (Table 2). However, rhPlGF-1 (5 or 10 µg/day) strongly synergized with rhG-CSF in increasing progenitor cell mobilization. Again, such a hematopoietic activity was dose-dependent, with the combined injection of 10 µg/day rhPlGF-1 plus rhG-CSF resulting in average increases of threefold for CFC frequency (p  .0001), fourfold for CFC absolute numbers (p  .001), and eightfold for LTC-IC absolute numbers (p  .001), as compared with rhG-CSF alone. Upon combination, the highest absolute CFC and LTC-IC counts were 12,000 and 1,700 per milliliter, respectively.3 S) v- ?& _- Z6 K/ ~' C

6 g8 |" D" i+ NTable 2. Hematopoietic mobilization in BALB/c mice upon treatment with rhG-CSF and rhPlGF-1
  U" h9 O, H4 A4 \" n0 V/ c( w1 \5 Y- B; [8 U4 S) ~% k/ Y
Although increasing the dose of rhPlGF-1 up to 15 µg/mouse per day failed to further improve rhG-CSF-elicited PBPC mobilization (data not shown), prolonging the combined rhPlGF-1/rhG-CSF administration up to 12 days resulted in a persistent and statistically significant enhancement of rhG-CSF-elicited PBPC mobilization (Table 3), with absolute counts of CFCs as high as 32,000 per milliliter.
0 s; x  r, O8 R4 S8 ^3 e" d
$ ^" a2 v' F, Z, |- wTable 3. Effects of a 12-day mobilization with rhG-CSF alone or in combination with rhPlGF-1 in BALB/c mice
& o) g/ ^; h) `! B- o  X+ @1 m$ f& l5 D9 g& I! D% w
To determine whether cells mobilized by rhPlGF-1 plus rhG-CSF could rescue irradiated animals, BALB/c mice were lethally irradiated (850 cGy) and injected with blood MNCs obtained from syngeneic donors that had been treated for 5 days with PBS/MSA, rhG-CSF (10 µg/day), or rhPlGF-1 (10 µg/day) plus rhG-CSF (10 µg/day). There was no survival in the irradiation control group (n = 10) or in the group (n = 10) injected with 5 x 105 PBS/MSA MNCs (Fig. 1). Injection of 2 x 105 cells mobilized by rhG-CSF or rhPlGF-1/rhG-CSF rescued 13% and 37% of recipient mice (n = 15 per group, p = .2), respectively. Injection of 5 x 105 cells mobilized by rhG-CSF or rhPlGF-1/rhG-CSF rescued 41% and 87% of recipient mice (n = 15 per group, p = .08), respectively. Although survivals of mice rescued by rhG-CSF- or rhPlGF-1/rhG-CSF-mobilized cells were not statistically different, a trend in favor of higher survival of mice rescued by rhPlGF-1/rhG-CSF-mobilized cells was evident at both cell dose levels.
; q0 X8 z' X( N! ?4 ?0 d/ Y8 B% H7 `2 i9 {* e& {
Figure 1. Radioprotective activity of recombinant human placental growth factor-1 (rhPlGF-1)/recombinant human granulocyte colony-stimulating factor (rhG-CSF)-mobilized cells. Survival of lethally irradiated (850 cGy) BALB/c mice at 60 days after transplantation with blood mononuclear cells (MNCs) from PBS/MSA-, rhG-CSF-, or rhPlGF-1/rhG-CSF-treated mice. Donor mice were treated once daily for 5 days with phosphate-buffered saline/mouse serum albumin (PBS/MSA), rhG-CSF (10 µg/mouse), or rhPlGF-1 (10 µg/mouse) plus rhG-CSF (10 µg/mouse). Control mice received irradiation only. Donor MNCs (2 x 105 or 5 x 105 cells per mouse) were injected into recipient mice via the tail vein 2¨C3 hours after irradiation. , irradiation only; , MNCs (5 x 105 cells per mouse) from PBS/MSA-treated mice;  MNCs (2 x 105 cells per mouse) from rhG-CSF-mobilized mice; , MNCs (2 x 105 cells per mouse) from rhPlGF-1/rhG-CSF-mobilized mice; , MNCs (5 x 105 cells per mouse) from rhG-CSF-mobilized mice; , MNCs (5 x 105 cells per mouse) from rhPlGF-1/rhG-CSF-mobilized mice.) T, I0 s$ \/ Y5 r6 T. {- g

$ x( O1 \- t" E$ S% jPlGF and MMP-9 Plasma Levels
' h1 {' c; E" @) v6 G
( l! v8 I- t/ v$ Z' _Under our experimental conditions, failure of rhPlGF-1 alone to mobilize PBPCs was associated with PlGF plasma levels' peaking 2 hours after cytokine injection and declining to undetectable levels within 24 hours (Fig. 2A). PlGF plasma levels achieved after i.p. administration might explain the lack of any mobilizing activity using rhPlGF-1 as a single agent. However, rhPlGF-1 strongly synergized with rhG-CSF in mobilizing PBPCs, thus allowing the hypothesis that the combined mobilization therapy may eventually enhance protease release/activation. Because MMP-9 has a critical role in PBPC mobilization by either releasing soluble Kit ligand (sKitL) , we quantified MMP-9 plasma levels in BALB/c mice receiving different mobilization regimens (Fig. 2B). Whereas rhPlGF-1 alone had a modest effect on MMP-9 plasma levels (p = .2), treatment with rhG-CSF or rhPlGF-1/rhG-CSF was associated with a twofold (p  .008) and threefold (p  .0001) increase of MMP-9 over baseline, respectively. As compared with rhG-CSF alone, the combined rhPlGF-1/rhG-CSF therapy induced significantly higher (p  .003) levels of MMP-9, thus suggesting that the effect at the protease level might represent the biological basis for the synergistic mobilizing activity of rhPlGF-1/rhG-CSF therapy.
6 |  G) B/ x" _* M
$ M) e4 O& D  \3 u7 c' \Figure 2. Plasma levels of human PlGF and mouse MMP-9 in BALB/c mice mobilized by recombinant human (rh)PlGF-1, (rh)G-CSF, or both. Cohorts of three BALB/c mice per group per experiment were injected i.p. for 4 days with control vehicle (PBS/MSA), rhPlGF-1 (10 µg/day), rhG-CSF (10 µg/day), or a combination of rhG-CSF (10 µg/day) and rhPlGF-1 (10 µg/day). Data are expressed as mean ¡À SEM. Statistical differences were evaluated using the Student's t test for unpaired data (two-tail). Combined data from three separate experiments are shown. (A): Plasma levels of human PlGF determined by enzyme-linked immunosorbent assay (ELISA). Collection of blood samples started 2 hours after the last injection of rhPlGF-1. (B): Plasma concentration of mouse MMP-9 determined by ELISA. Blood samples were collected 2 hours after the last injection of cytokine. *, p 0 L7 m; W7 f5 ~6 G4 E4 G) G/ Z

! {! q0 g4 J8 _% Y8 u2 A% ]Effects of rhPlGF-1 and rhG-CSF on Circulating WBCs, MNCs, and Platelets in Rhesus Monkeys
3 _7 ?) |- p; B( o! g# h$ O# Y
* J* ]+ t2 A* K( x) A' s2 qDue to the significant activity of rhPlGF-1/rhG-CSF in mice, we further explored the capability of rhPlGF-1 to mobilize PBPCs in nonhuman primates. Rhesus monkeys received four mobilization cycles at 6-week intervals. After a standard mobilization with rhG-CSF alone (100 µg/kg per day for 5 days) (cycle 1) , monkeys received rhG-CSF in combination with rhPlGF-1 at either 130 (cycle 2) or 260 µg/kg per day (cycle 3). A fourth cycle consisting of rhPlGF-1 alone (260 µg/kg per day) was finally administered (cycle 4). Because the 5-day treatment with rhPlGF-1 alone had no mobilization effect (data not shown), only the results observed in monkeys receiving rhG-CSF alone or in combination with rhPlGF-1 are reported here.
' u1 L' Q  r7 V/ y0 z" U6 D5 B: m7 ?0 G" u0 g
When administered alone, rhG-CSF induced an average increment of fivefold for WBCs (8,708 vs. 43,523, p  .0008) and threefold for MNCs (5,543 vs. 14,738, p  .04), whereas platelets (PLTs) were slightly reduced (Fig. 3A¨C3C). As compared with rhG-CSF alone, the combined rhPlGF-1/rhG-CSF therapy further increased day-5 WBCs at both dose levels (i.e., 130 ), whereas MNC and PLT values were not significantly changed (Fig. 3B, 3C). Five days after cessation of mobilization therapy, WBC, MNC, and PLT counts had returned to pretreatment levels.4 {, Q0 {4 L* u6 B( Q; A  |% H9 S! ^7 n

6 F: y6 s. H8 X- {Figure 3. Effects of rhPlGF-1 and/or rhG-CSF on WBC, MNC, and PLT counts. Rhesus monkeys (n = 4) received 5-day mobilization cycles at 6-week intervals. Mobilization was elicited at cycle 1 () by rhG-CSF alone (100 µg/kg per day, s.c.), at cycle 2 () by a combination of rhPlGF-1 (130 µg/kg, i.v.) plus rhG-CSF (100 µg/kg per day, s.c.), at cycle 3 () by a combination of rhPlGF-1 (260 µg/kg, i.v.) plus rhG-CSF (100 µg/kg per day, s.c.), and at cycle 4 (data not shown) by rhPlGF-1 alone (260 µg/kg, i.v.). Data are expressed as mean ¡À SEM. Statistical differences were evaluated using the Student's t test for paired data (two-tail). (A): WBC counts. The average pretreatment WBC counts per microliter of blood at cycles 1, 2, and 3 were 8,708 ¡À 1,299, 13,498 ¡À 2,757, and 8,370 ¡À 793, respectively. (B): MNC counts. The average pretreatment MNC counts per microliter of blood at cycles 1, 2, and 3 were 5,543 ¡À 973, 3,325 ¡À 519, and 4,975 ¡À 554, respectively. (C): PLT counts. The average pretreatment PLT counts per microliter of blood at cycles 1, 2, and 3 were 349,500 ¡À 11,637, 365,000 ¡À 31,507, and 355,500 ¡À 40,556, respectively. *, p 2 k+ ~3 c& l( d; @1 @/ l

; ^; z+ Y' A! v! {CFC Mobilizing Effects of rhPlGF-1 and rhG-CSF in Rhesus Monkeys
- d/ p5 q! z) f  K" m; Y* \4 Q8 X9 _* h) ~( ^$ e5 a: L
As compared with baseline, the mean frequency of blood CFCs at peak was increased by 16-fold (p  .0009), 33-fold (p  .0002), and 41-fold (p  .0001) under rhG-CSF alone, rhG-CSF plus rhPlGF-1 at 130 µg/kg per day, and rhG-CSF plus rhPlGF-1 at 260 µg/kg per day, respectively (Table 4). The combined rhPlGF-1/rhG-CSF treatment induced a significant increase of CFC frequencies over those detected under rhG-CSF alone, with increments of twofold (p  .004) and 2.5-fold (p  .004) when rhPlGF-1 was injected at 130 and 260 µg/kg per day, respectively (Table 4). Five days after cessation of treatment, CFCs had almost returned to baseline values.
$ A$ L0 r. q( k7 ^: H5 T  r
# {* Q# Z" m& ITable 4. Hematopoietic mobilization in rhesus monkeys upon treatment with rhG-CSF and rhPlGF-1
! ~" f0 f* M) W
0 Z* O- s2 `# k; zAbsolute numbers of circulating CFCs in blood were calculated as a function of the frequency of CFCs multiplied by the total number of MNCs per milliliter of blood. Whereas under steady-state conditions 141 ¡À 26 CFCs per milliliter of blood could be detected, this value was increased by 72-fold (p  .01), 322-fold (p  .001), and 388-fold (p  .004) under rhG-CSF alone, rhG-CSF plus rhPlGF-1 at 130 µg/kg per day, and rhG-CSF plus rhPlGF-1 at 260 µg/kg per day, respectively (Fig. 4A). Thus, rhG-CSF-elicited mobilization of CFCs per milliliter of blood was increased by fourfold (p  .006) and fivefold (p  .006) under rhG-CSF plus rhPlGF-1 at 130 and 260 µg/kg per day, respectively.
) S; {3 M' o' s' \& b  G- G+ S" H' g# ]; i3 X
Figure 4. Absolute values of circulating CFCs and HPP-CFCs in monkeys mobilized with recombinant human placental growth factor-1 (rhPlGF-1) and/or recombinant human granulocyte colony-stimulating factor (rhG-CSF). Rhesus monkeys (n = 4) received 5-day mobilization cycles at 6-week intervals. Mobilization was elicited at cycle 1 () by rhG-CSF alone (100 µg/kg per day, s.c.), at cycle 2 () by a combination of rhPlGF-1 (130 µg/kg, i.v.) plus rhG-CSF (100 µg/kg per day, s.c.), at cycle 3 () by a combination of rhPlGF-1 (260 µg/kg, i.v.) plus rhG-CSF (100 µg/kg per day, s.c.), and at cycle 4 (data not shown) by rhPlGF-1 alone (260 µg/kg, i.v.). Data are expressed as mean ¡À SEM derived from quadruplicate cultures on samples from each animal at each time point. Statistical differences were evaluated using the Student's t test for paired data (two-tail). (A): Absolute values per milliliter of blood of circulating CFCs. CFCs include granulocyte-macrophage CFC (CFU-GM), erythroid burst-forming unit (BFU-E), and multipotent CFC (CFU-Mix). The absolute number of circulating CFCs in blood is a function of the frequency of CFCs multiplied by the total number of mononuclear cells (MNCs) per milliliter of blood. The average CFC count in control monkeys was 141 ¡À 26 per milliliter of blood. (B): HPP-CFCs were defined as macroscopically visible colonies of more than 2 mm in diameter of compact colony growth and were scored after 28 days of incubation from methylcellulose cultures. Absolute values per milliliter of blood of circulating HPP-CFCs. The absolute number of circulating HPP-CFCs in blood is a function of the frequency of HPP-CFCs multiplied by the total number of MNCs per milliliter of blood. The average HPP-CFC count in control monkeys was 32 ¡À 7 per milliliter of blood. ¡ã, p
2 }8 F% ]! L; Z" e- a/ c' l6 F0 n$ B4 k5 ?4 X# _" ^
HPP-CFC Mobilizing Effects of rhPlGF-1 and rhG-CSF in Rhesus Monkeys
6 {& {* M9 H4 N
  h0 \5 g6 N& W5 d, c' \In addition to CFCs, macroscopically visible colonies of more than 2 mm in diameter of compact growth (HPP-CFCs) were assessed in methylcellulose culture scored after 28 days of incubation . On day 5 of mobilization, HPP-CFC frequency was increased by 6-fold (p  .03), 12-fold (p  .002), and 16-fold (p  .01) under rhG-CSF alone, rhG-CSF plus rhPlGF-1 at 130 µg/kg per day, and rhG-CSF plus rhPlGF-1 at 260 µg/kg per day, respectively (Table 4). The combined rhPlGF-1/rhG-CSF treatment induced a significant increase of HPP-CFC frequencies over those detected under rhG-CSF alone, with increments of twofold (p  .001) and 2.5-fold (p  .001) when rhPlGF-1 was injected at 130 and 260 µg/kg per day, respectively (Table 4).& q7 f8 B+ i! v( t
( `( P1 D5 D/ ~3 j9 G
Under steady-state conditions, 32 ¡À 7 HPP-CFCs were detected per milliliter of blood. This value was increased by 50-fold (p  .002), 280-fold (p  .0007), and 337-fold (p  .0007) under rhG-CSF alone, rhG-CSF plus rhPlGF-1 at 130 µg/kg per day, and rhG-CSF plus rhPlGF-1 at 260 µg/kg per day, respectively (Fig. 4B). The peak levels of HPP-CFCs per milliliter of blood induced by rhG-CSF were increased by sixfold (p  .001) and sevenfold (p  .001) upon administration of rhG-CSF plus rhPlGF-1 at 130 and 260 µg/kg per day, respectively (Fig. 4B).
; G; L! D4 a! X5 V1 [2 q& T4 p
6 \6 Y- R0 _; W, j* cLTC-IC Mobilizing Effects of rhPlGF-1 and rhG-CSF in Rhesus Monkeys
1 _* C/ S7 L4 @2 P0 W4 d8 p5 P$ G7 K
To further evaluate the mobilizing activity of the combined rhPlGF-1/rhG-CSF treatment, we investigated the frequency and the absolute numbers per milliliter of blood of the primitive hematopoietic progenitors capable of forming colonies in long-term culture (LTC-IC). As compared with rhG-CSF alone, the combined administration of rhG-CSF and rhPlGF-1 at 130 µg/kg per day increased the mean frequency of LTC-ICs by 14-fold (i.e., from 1 LTC-IC in 83,237 up to 1 in 5,829  MNCs) (Fig. 5A). Increasing the dose of rhPlGF-1 at 260 µg/kg per day did not result in a further increase of LTC-IC frequency (data not shown). As compared with rhG-CSF alone, the absolute numbers of circulating LTC-ICs detected under rhG-CSF plus rhPlGF-1 at 130 and 260 µg/kg per day were increased by 12-fold (p  .006) and 15-fold (p  .007), respectively (Fig. 5B).
- X, ~: q. }0 A" G  ^7 ?
: x5 |" [- [8 q& B* pFigure 5. Frequency and absolute values of circulating LTC-ICs in monkeys mobilized with recombinant human (rh)PlGF-1 and/or rhG-CSF. Rhesus monkeys (n = 4) received 5-day mobilization cycles at 6-week intervals. Mobilization was elicited at cycle 1 () by rhG-CSF alone (100 µg/kg per day, s.c.), at cycle 2 () by a combination of rhPlGF-1 (130 µg/kg, i.v.) plus rhG-CSF (100 µg/kg per day, s.c.), at cycle 3 () by a combination of rhPlGF-1 (260 µg/kg, i.v.) plus rhG-CSF (100 µg/kg per day, s.c.), and at cycle 4 (data not shown) by rhPlGF-1 alone (260 µg/kg, i.v.). Data are expressed as mean ¡À SEM. Statistical differences were evaluated using the Student's t test for paired data (two-tail). (A): Frequency of circulating LTC-ICs (per 105 mononuclear cells ). The frequency of LTC-ICs was assayed under limiting dilution conditions using the murine M2-10B4 cell line (kindly provided by Dr. C. Eaves, Terry Fox Laboratory, Vancouver, BC, Canada) as stromal layer. Blood samples were collected on day 5 of mobilization therapy. Serial dilutions of test cells (2 x 105 to 3 x 10) were cultured for 5 weeks, and 16¨C22 replicates were plated for each test cell dose. After 5 weeks, nonadherent and adherent cells from individual wells were assayed for clonogenic cells, and the LTC-IC frequencies were calculated using Poisson statistics and the method of maximum likelihood. (B): Absolute values per milliliter of blood of circulating LTC-ICs. The absolute number of circulating LTC-ICs in blood is a function of the frequency of LTC-ICs multiplied by the total number of MNCs per milliliter of blood. The average LTC-IC count in control monkeys was 21 ¡À 6 per milliliter of blood. *, p
9 J! E% K" O( U6 m! S' i5 i. j5 o6 |
Toxicity2 ]' t" F5 X  [

/ M0 a, O3 t  D5 y  ~; ~No specific toxicity was associated with the administration of rhPlGF-1 alone or in combination with rhG-CSF. Mobilization therapy did not affect body weight, body temperature, or blood pressure. In monkeys receiving rhPlGF-1 alone or rhPlGF-1/rhG-CSF, no significant changes in serum values of electrolytes, blood urea nitrogen, creatinine, hepatic aminotransferases, bilirubin, alkaline phosphatase, coagulation profiles, serum chemistry, or urine analysis could be detected.
0 V' ~/ K0 f9 B1 L" W* {5 }8 o& K7 U1 w0 P  `
DISCUSSION
& y* p' i! w$ V- L9 S3 d6 R. f  R: ~2 S9 ]; V
Attempts to enhance the quality, quantity, and kinetics of cytokine-induced mobilization of PBPCs have been pursued by using a variety of cytokine combination protocols, including rhGM-CSF plus rhG-CSF . In this scenario, rhPlGF-1 might represent a new cytokine to be exploited due to its potent enhancing effect on PBPC mobilization when used in combination with rhG-CSF.
) I( H1 F6 R, b$ L5 d0 M
4 i8 W0 A- L6 @, z$ H( ABy using two different animal models (i.e., BALB/c mice and rhesus monkeys) that allow simulation of PBPC mobilization as occurs in a clinical situation, we demonstrate herein that rhPlGF-1 synergizes with rhG-CSF in enhancing the frequencies and absolute numbers of a broad spectrum of circulating hematopoietic progenitors, including committed CFCs, HPP-CFCs, primitive LTC-ICs, and radioprotective cells. In BALB/c mice, rhPlGF-1 enhanced rhG-CSF-elicited mobilization of CFCs and LTC-ICs per milliliter of blood by fourfold and eightfold, respectively. In rhesus monkeys, rhPlGF-1 enhanced rhG-CSF-elicited mobilization of CFCs, HPP-CFCs, and LTC-ICs per milliliter of blood by 5-fold, 7-fold, and 15-fold, respectively. Our data significantly extend previous observations in myelosuppressed mice showing that injection of an adenovector expressing PlGF enhances early phases of marrow recovery directly by recruitment of VEGFR-1  marrow repopulating cells, whereas in later stages PlGF indirectly supports hematopoiesis through release of sKitL mediated by MMP-9 .
' ]; r" `7 }( ~0 U) z+ G2 V" g& a0 s2 i' F
  W' L, ?5 r1 C5 ^( u* nThe lack of WBC and progenitor cell release into the bloodstream under rhPlGF-1 alone which we observed in our model systems is in contrast with previous data .
" E* I6 A' _6 ^3 y. B
& [. R: c" x) O' H  ?! JThe lower dose of rhPlGF-1 tested in monkeys (i.e., 130 µg/kg per day) was calculated from mice studies using an appropriate conversion factor. The monkey dose was biologically equivalent to the dose of rhPlGF-1 (10 µg/day) which induced a maximal enhancement of rhG-CSF-elicited PBPC mobilization in BALB/c mice , translating the results of the combined rhPlGF-1/rhG-CSF therapy from monkeys to humans would allow us to predict that at least a 1-log increase of committed and primitive progenitors should be achievable both in cancer patients and normal donors in the absence of any additional toxicity./ B0 q2 |$ y1 ?1 t( P" W7 w
" d: ~+ g" K9 H8 y% v
According to our study design in monkeys, the second and the third mobilization cycles (i.e., rhPlGF-1 plus rhG-CSF) were compared with the first mobilization cycle (i.e., rhG-CSF alone), with each mobilization treatment being separated by a 6-week washout period. For each animal, the kinetic of mobilization after cycle 1 served as intramonkey control to assess the mobilization after cycles 2 and 3. This study design was aimed at preventing the effects the interanimal variability and was based on previous data showing that repeated mobilization cycles have no enhancing or detrimental effect on hematopoietic mobilization provided that an adequate washout period was included between each mobilization procedure . Based on this hypothesis, we did not include a control group receiving rhG-CSF alone at cycles 2 and 3.
; S) B/ F. t1 N2 C+ K! C0 f9 p% M) P) ~# u; Z  `
The PlGF receptor, VEGFR-1, is expressed by endothelial cells but is also present on inflammatory cells such as monocytes and macrophages . The application of VEGFR-1 ligands for hematopoietic stem cell recruitment and mobilization could be useful not only in patients undergoing SCT but also in patients with suppressed bone marrow function after irradiation and chemotherapy. However, problems could arise with systemic VEGFR-1 therapies in that VEGFR-1 stimulation of neovascularization could also accelerate retinopathy, plaque formation, or possibly tumor growth. All these issues should be carefully addressed in future clinical studies aimed at exploring the hematopoietic modulating effects of rhPlGF-1.
$ h. E4 }3 A6 q: V$ f- P$ W7 B+ [, }8 D
Our data demonstrate that rhPlGF-1 significantly enhances rhG-CSF-elicited hematopoietic mobilization and provide a preclinical rationale for evaluating the potential clinical benefit of rhPlGF-1 in conjunction with rhG-CSF to mobilize PBPCs for application in peripheral blood SCT, gene therapy, and/or immunotherapy. A dose-finding study aimed at identifying the dose of rhPlGF-1 capable of improving rhG-CSF-elicited mobilization is planned in patients with cancer." ^- Q( ~% |! e  I. L) P$ L

8 j2 B+ G0 q$ k! U- lDISCLOSURES
8 |6 H! ~! z+ v9 p" I4 t$ D/ i/ n. U( T$ d9 d( u
The authors indicate no potential conflicts of interest.; w; d9 Y+ G4 E" y/ ?

2 W: l/ Y  l/ p. Y0 jACKNOWLEDGMENTS/ I- ]5 J  X# W: Q9 z
" ]# u" A! u5 I/ a& k2 U0 Q: c
This work was supported in part by grants from Ministero dell'Istruzione, dell'Universit¨¤ e della Ricerca (Rome), Ministero della Salute (Rome), and Michelangelo Foundation for Advances in Cancer Research and Treatment (Milan, Italy). We thank Dr. A. Mion (Geymonat S.p.A.) for the kind gift of rhPlGF-1.: L4 g1 i, M) e0 ?2 u6 l# R
          【参考文献】
/ f  @* F3 J1 U+ S1 l7 K. d  x
4 H+ u& [) q/ ?6 F6 f! b
6 w0 j. p( s. h& b! r6 k- TBarlogie B. High-dose therapy and innovative approaches to treatment of multiple myeloma. Semin Hematol 2001;38:21¨C27." v* a5 V: f6 Z9 Q
% W' g- T+ B! c1 u
Ferme C, Mounier N, Divine M et al. Intensive salvage therapy with high-dose chemotherapy for patients with advanced Hodgkin's disease in relapse or failure after initial chemotherapy: Results of the Groupe d'Etudes des Lymphomes de l'Adulte H89 Trial. J Clin Oncol 2002;20:467¨C475.$ t+ G8 l) A) r
1 t' i1 k' d2 S9 k. k
Gianni AM. High-dose chemotherapy and autotransplants: A time for guidelines. Ann Oncol 1997;8:933¨C935.
. l& u$ a; P, a% ?3 y
1 x" X( T7 W/ {- g- x- ZGianni AM, Bregni M, Siena S et al. High-dose chemotherapy and autologous bone marrow transplantation compared with MACOP-B in aggressive B-cell lymphoma. N Engl J Med 1997;336:1290¨C1297.
" O3 R% E7 q5 c9 z  s' \+ [* Y
, s( i7 S. e" `: TAppelbaum FR. Choosing the source of stem cells for allogeneic transplantation: No longer a peripheral issue. Blood 1999;94:381¨C383.2 s/ x. l+ e2 O0 n: V" Q

! Q$ s! V3 o. t: F. _" Q( bAversa F, Tabilio A, Terenzi A et al. Successful engraftment of T-cell-depleted haploidentical "three-loci" incompatible transplants in leukemia patients by addition of recombinant human granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells to bone marrow inoculum. Blood 1994;84:3948¨C3955.
4 i3 ?0 U2 g  P5 t( w- w
  T$ j( x1 }+ P1 D( KBensinger WI, Martin PJ, Storer B et al. Transplantation of bone marrow as compared with peripheral-blood cells from HLA-identical relatives in patients with hematologic cancers. N Engl J Med 2001;344:175¨C181.
$ |5 F" Y& n$ v" {
# j6 D. k. h7 v# \8 ?5 [0 a6 lDemirer T, Bensinger WI, Buckner CD. Peripheral blood stem cell mobilization for high-dose chemotherapy. J Hematother 1999;8:103¨C113.7 Z2 M# e. G) {& d* G6 N
4 @9 ?9 E$ G. l+ x9 [
Haas R, Mohle R, Fruhauf S et al. Patient characteristics associated with successful mobilizing and autografting of peripheral blood progenitor cells in malignant lymphoma. Blood 1994;83:3787¨C3794.1 n! b6 W" @/ _; i/ C
  G9 s* q: n  t6 d: D9 f. f( X
Dreger P, Kloss M, Petersen B et al. Autologous progenitor cell transplantation: Prior exposure to stem cell-toxic drugs determines yield and engraftment of peripheral blood progenitor cell but not of bone marrow grafts. Blood 1995;86:3970¨C3978.5 \+ G  f! v0 m7 @; L; N

" n+ E# D$ Z! U- J) `; ^Tarella C, Di Nicola M, Caracciolo D et al. High-dose ara-C with autologous peripheral blood progenitor cell support induces a marked progenitor cell mobilization: An indication for patients at risk for low mobilization. Bone Marrow Transplant 2002;30:725¨C732." D) w! A1 W! N
; n1 x" M: N7 p5 V+ F9 A3 A# N  `
Weaver CH, Hazelton B, Birch R et al. An analysis of engraftment kinetics as a function of the CD34 content of peripheral blood progenitor cell collections in 692 patients after the administration of myeloablative chemotherapy. Blood 1995;86:3961¨C3969.
5 g' N2 v! q: r; O" k) o5 J5 F, F3 m# Q8 I: f9 \
Carlo-Stella C, Di Nicola M, Magni M et al. Defibrotide in combination with granulocyte colony-stimulating factor significantly enhances the mobilization of primitive and committed peripheral blood progenitor cells in mice. Cancer Res 2002;62:6152¨C6157.
7 f' [6 r; \& t* W
8 a( Z# s! B% e9 C8 P( O, C5 XCraddock CF, Nakamoto B, Andrews RG et al. Antibodies to VLA4 integrin mobilize long-term repopulating cells and augment cytokine-induced mobilization in primates and mice. Blood 1997;90:4779¨C4788.0 ^2 |6 }6 F6 u. `/ g. s

, I! I2 `) ]4 ^. D; PKing AG, Horowitz D, Dillon SB et al. Rapid mobilization of murine hematopoietic stem cells with enhanced engraftment properties and evaluation of hematopoietic progenitor cell mobilization in rhesus monkeys by a single injection of SB-251353, a specific truncated form of the human CXC chemokine GRObeta. Blood 2001;97:1534¨C1542.$ J- y  F! z5 [; C& j; o
7 ]: B4 R: @( F9 Q
Liles WC, Broxmeyer HE, Rodger E et al. Mobilization of hematopoietic progenitor cells in healthy volunteers by AMD3100, a CXCR4 antagonist. Blood 2003;102:2728¨C2730.9 V2 o2 Y* i: @0 r0 r7 Y

5 A# e  B7 l) R9 aBishop MR, Jackson JD, O'Kane-Murphy B et al. Phase I trial of recombinant fusion protein PIXY321 for mobilization of peripheral-blood cells. J Clin Oncol 1996;14:2521¨C2526.
  i1 P) y7 h- @: H- P, D% B0 T
4 q# [6 k; T' W1 b4 cBrasel K, McKenna HJ, Charrier K et al. Flt3 ligand synergizes with granulocyte-macrophage colony-stimulating factor or granulocyte colony-stimulating factor to mobilize hematopoietic progenitor cells into the peripheral blood of mice. Blood 1997;90:3781¨C3788." P  h1 U8 J1 @

3 l$ z  \: y# r+ ]- fKoc ON, Gerson SL, Cooper BW et al. Randomized cross-over trial of progenitor-cell mobilization: High-dose cyclophosphamide plus granulocyte colony-stimulating factor (G-CSF) versus granulocyte-macrophage colony-stimulating factor plus G-CSF. J Clin Oncol 2000;18:1824¨C1830.
- b; A8 v: [/ h8 P! s9 g) u* j# w
7 E7 C2 P' N( _7 x/ P$ p8 O  B, U2 ~8 aRosenfeld CS, Bolwell B, LeFever A et al. Comparison of four cytokine regimens for mobilization of peripheral blood stem cells: IL-3 alone and combined with GM-CSF or G-CSF. Bone Marrow Transplant 1996;17:179¨C183.1 s4 @2 ~& U' J# ?+ {
! A2 Z. `+ {% U; @# h2 m  ]# ]7 L! r
Shpall EJ, Wheeler CA, Turner SA et al. A randomized phase 3 study of peripheral blood progenitor cell mobilization with stem cell factor and filgrastim in high-risk breast cancer patients. Blood 1999;93:2491¨C2501.
" R: I; y1 V9 l/ D' B2 x' }7 i6 t6 h  D5 x
Stiff P, Gingrich R, Luger S et al. A randomized phase 2 study of PBPC mobilization by stem cell factor and filgrastim in heavily pretreated patients with Hodgkin's disease or non-Hodgkin's lymphoma. Bone Marrow Transplant 2000;26:471¨C481.
0 j3 n9 v, s) \1 A* C6 ~
. }5 n% R3 \& M. fTo LB, Bashford J, Durrant S et al. Successful mobilization of peripheral blood stem cells after addition of ancestim (stem cell factor) in patients who had failed a prior mobilization with filgrastim (granulocyte colony-stimulating factor) alone or with chemotherapy plus filgrastim. Bone Marrow Transplant 2003;31:371¨C378.; V5 P# H/ A$ ^% b% c* ?- n

! |, t9 F9 O5 Z/ L! B4 n6 XFruehauf S, Seggewiss R. It's moving day: Factors affecting peripheral blood stem cell mobilization and strategies for improvement/ G7 `6 J2 L" A1 L+ v7 A; T( |
& H9 S$ y4 h  _# X& s
Kessinger A, Sharp JG. The whys and hows of hematopoietic progenitor and stem cell mobilization. Bone Marrow Transplant 2003;31:319¨C329.( ~$ Q3 C  s8 w* s
6 r. h$ o6 b0 h" F# d
Liles WC, Rodger E, Broxmeyer HE et al. Augmented mobilization and collection of CD34  hematopoietic cells from normal human volunteers stimulated with granulocyte-colony-stimulating factor by single-dose administration of AMD3100, a CXCR4 antagonist. Transfusion 2005;45:295¨C300.
. w9 o- ~  D! }# C0 `
* I/ X+ A: X: x) q( D* O$ bBroxmeyer HE, Orschell CM, Clapp DW et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med 2005;201:1307¨C1318.
/ S5 z0 Q$ h8 N6 C- T( f0 U: r# U1 g9 o6 K3 Y
Devine SM, Flomenberg N, Vesole DH et al. Rapid mobilization of CD34  cells following administration of the CXCR4 antagonist AMD3100 to patients with multiple myeloma and non-Hodgkin's lymphoma. J Clin Oncol 2004;22:1095¨C1102.
, S- N+ J1 f* B4 s: a/ m# z; I4 {0 m" u3 Q3 e, f/ g
Carlo-Stella C, Di Nicola M, Milani R et al. Use of recombinant human growth hormone (rhGH) plus recombinant human granulocyte colony-stimulating factor (rhG-CSF) for the mobilization and collection of CD34  cells in poor mobilizers. Blood 2004;103:3287¨C3295.
; K" l% q( ?4 }8 L; e
5 e0 F; U) o# hCao Y, Ji WR, Qi P et al. Placenta growth factor: Identification and characterization of a novel isoform generated by RNA alternative splicing. Biochem Biophys Res Commun 1997;235:493¨C498.
$ B0 o4 i& z/ `5 k! C: \* {: D2 v; K% B( h2 ^- q
Maglione D, Guerriero V, Viglietto G et al. Isolation of a human placenta cDNA coding for a protein related to the vascular permeability factor. Proc Natl Acad Sci U S A 1991;88:9267¨C9271." P; c2 f* p. i# W0 ?6 \( B: R$ b( f

- ~' @6 [1 y" _! C# J/ o" `Maglione D, Guerriero V, Viglietto G et al. Two alternative mRNAs coding for the angiogenic factor, placenta growth factor (PlGF), are transcribed from a single gene of chromosome 14. Oncogene 1993;8:925¨C931.
/ T7 P3 q" L5 C& M9 n$ k! f, c4 S, x
Yang W, Ahn H, Hinrichs M et al. Evidence of a novel isoform of placenta growth factor (PlGF-4) expressed in human trophoblast and endothelial cells. J Reprod Immunol 2003;60:53¨C60.
9 w8 B- X8 {3 P+ d
0 m6 U+ x# z5 w! UCarmeliet P, Moons L, Luttun A et al. Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasation in pathological conditions. Nat Med 2001;7:575¨C583.8 p: ^' |* b0 P

* e0 m. \; D4 l( O+ B# ^: `Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003;9:669¨C676.2 V0 r* ]4 P4 W* ]" w0 C# }* p5 j
& H1 w9 x( E7 D: }) B# f
Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med 2003;9:702¨C712.
5 ~! D: c% F1 q6 Q7 w" Z0 f; d; I6 h7 l  g8 o0 p1 _4 I& Y
Hattori K, Heissig B, Wu Y et al. Placental growth factor reconstitutes hematopoiesis by recruiting VEGFR1( ) stem cells from bone-marrow microenvironment. Nat Med 2002;8:841¨C849.% T2 {2 s% k' B
, H  a$ \6 `/ g- z8 m$ C* V
UKCCCR guidelines for the welfare of animals in experimental neoplasia. Br J Cancer 1988;58:109¨C113.( R' `! R; _% k  Q

- \3 Z' u, |+ {# T8 iMolineux G, McCrea C, Yan XQ et al. Flt-3 ligand synergizes with granulocyte colony-stimulating factor to increase neutrophil numbers and to mobilize peripheral blood stem cells with long-term repopulating potential. Blood 1997;89:3998¨C4004.
' b2 T+ \. q7 X" k0 B
; \5 {( Y6 t/ b' V; e6 nSudo Y, Shimazaki C, Ashihara E et al. Synergistic effect of FLT-3 ligand on the granulocyte colony-stimulating factor-induced mobilization of hematopoietic stem cells and progenitor cells into blood in mice. Blood 1997;89:3186¨C3191.3 l9 S% h- M. _' d& }7 S6 r9 l
, c$ d0 y8 N5 ?
Carlo-Stella C, Di Nicola M, Milani R et al. Age- and irradiation-associated loss of bone marrow hematopoietic function in mice is reversed by recombinant human growth hormone. Exp Hematol 2004;32:171¨C178.
! Q) n  b" |) y& }! N( _  a$ Z8 `5 Y/ [/ S. t9 K4 P# e
Humphries RK, Eaves AC, Eaves CJ. Characterization of a primitive erythropoietic progenitor found in mouse marrow before and after several weeks in culture. Blood 1979;53:746¨C763.
; o, r7 A& Y: L
/ g8 M  Y6 y4 ~* aMoore KA, Ema H, Lemischka IR. In vitro maintenance of highly purified, transplantable hematopoietic stem cells. Blood 1997;89:4337¨C4347.
8 E! X& H; [$ W; O  _  @
; N, r4 u% {# r& zSutherland HJ, Eaves CJ, Eaves AC et al. Characterization and partial purification of human marrow cells capable of initiating long-term hematopoiesis in vitro. Blood 1989;74:1563¨C1570.
3 i2 F9 h% U# B/ T$ q' `) w% B
. X. f( x! I2 B, X; ~, i! g( @Pruijt JF, van Kooyk Y, Figdor CG et al. Anti-LFA-1 blocking antibodies prevent mobilization of hematopoietic progenitor cells induced by interleukin-8. Blood 1998;91:4099¨C4105.
6 o) Y8 L9 v, m" ~6 R" o% y; E- _# A
Carlo-Stella C, Di Nicola M, Longoni P et al. Mobilization of primitive and committed hematopoietic progenitors in nonhuman primates treated with defibrotide and recombinant human granulocyte colony-stimulating factor. Exp Hematol 2004;32:68¨C75.
1 d! f5 V+ X& v: C5 r$ J& P9 L  t; b/ H$ h" O( i, j
MacVittie TJ, Farese AM, Davis TA et al. Myelopoietin, a chimeric agonist of human interleukin 3 and granulocyte colony-stimulating factor receptors, mobilizes CD34  cells that rapidly engraft lethally x-irradiated nonhuman primates. Exp Hematol 1999;27:1557¨C1568.8 X5 }. W: l) P" T/ E# j

( ?; o+ ]! {7 ?" vFreireich EJ, Gehan EA, Rall DP et al. Quantitative comparison of toxicity of anticancer agents in mouse, rat, hamster, dog, monkey, and man. Cancer Chemother Rep 1966;50:219¨C244.
* B  q$ h% _- G: z
0 I$ ]5 H6 c. ]* \) GLemieux ME, Rebel VI, Lansdorp PM et al. Characterization and purification of a primitive hematopoietic cell type in adult mouse marrow capable of lymphomyeloid differentiation in long-term marrow "switch" cultures. Blood 1995;86:1339¨C1347.& ^! d- M; _5 g" P. @& D
' U. d' O$ n+ l# O2 q
Sutherland HJ, Eaves CJ, Lansdorp PM et al. Differential regulation of primitive human hematopoietic cells in long-term cultures maintained on genetically engineered murine stromal cells. Blood 1991;78:666¨C672.3 L0 |+ M: M* k' O' @- Y

! J& s- X. O  ZHeissig B, Hattori K, Dias S et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell 2002;109:625¨C637.
2 p4 ^' A1 B9 I+ f  V3 T% s& H8 l! V$ z% p
McQuibban GA, Butler GS, Gong JH et al. Matrix metalloproteinase activity inactivates the CXC chemokine stromal cell-derived factor-1. J Biol Chem 2001;276:43503¨C43508.
8 r; f3 ~  q5 M) _) V7 t- N" E2 S$ w, x; o" _6 F
Pelus LM, Bian H, King AG et al. Neutrophil-derived MMP-9 mediates synergistic mobilization of hematopoietic stem and progenitor cells by the combination of G-CSF and the chemokines GRObeta/CXCL2 and GRObetaT/CXCL2delta4. Blood 2004;103:110¨C119.7 {* {, I5 y- k& d( x- c
: S2 \- c8 Z, L* f
Petit I, Szyper-Kravitz M, Nagler A et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol 2002;3:687¨C694.5 M. t, A& J, ?- J

* i+ t( K& {) k' d6 FEriksson U, Alitalo K. VEGF receptor 1 stimulates stem-cell recruitment and new hope for angiogenesis therapies. Nat Med 2002;8:775¨C777.; ~1 s5 I6 }5 P7 a7 c, R

8 w8 K/ Y8 O& CFejer G, Szalay K, Gyory I et al. Adenovirus infection dramatically augments lipopolysaccharide-induced TNF production and sensitizes to lethal shock. J Immunol 2005;175:1498¨C1506.
9 H  O& B' l$ C3 X8 w$ L3 [) D6 [0 _4 d6 E1 _3 @/ t! u
Levesque JP, Hendy J, Takamatsu Y et al. Disruption of the CXCR4/CXCL12 chemotactic interaction during hematopoietic stem cell mobilization induced by GCSF or cyclophosphamide. J Clin Invest 2003;111:187¨C196.8 B9 R5 ~. y  U

' ]$ f: E* r, i* c: M# D. nLevesque JP, Hendy J, Winkler IG et al. Granulocyte colony-stimulating factor induces the release in the bone marrow of proteases that cleave c-KIT receptor (CD117) from the surface of hematopoietic progenitor cells. Exp Hematol 2003;31:109¨C117.
( _+ g- z: y+ g9 H7 V( i5 b5 X* v; X  e4 n4 Y: t$ S9 u) X# ]! c" \$ \
Levesque JP, Takamatsu Y, Nilsson SK et al. Vascular cell adhesion molecule-1 (CD106) is cleaved by neutrophil proteases in the bone marrow following hematopoietic progenitor cell mobilization by granulocyte colony-stimulating factor. Blood 2001;98:1289¨C1297.
2 k0 ]  ^$ ]5 c$ U$ m8 |6 Q
8 R+ V- O4 Z$ L" j, a7 r) iShi PA, Pomper GJ, Metzger ME et al. Assessment of rapid remobilization intervals with G-CSF and SCF in murine and rhesus macaque models. Transfusion 2001;41:1438¨C1444.* X: v: d& N/ X$ V

8 U& J" `: K2 h4 ]Gerber HP, Malik AK, Solar GP et al. VEGF regulates haematopoietic stem cell survival by an internal autocrine loop mechanism. Nature 2002;417:954¨C958.3 k. Y# l" e8 `- m! ]5 D

- Y& S5 ?8 J/ e& w5 M* l! N( C7 fLuttun A, Tjwa M, Moons L et al. Revascularization of ischemic tissues by PlGF treatment, and inhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1. Nat Med 2002;8:831¨C840.

Rank: 2

积分
77 
威望
77  
包包
1730  
沙发
发表于 2015-5-28 14:19 |只看该作者
干细胞之家是不错的网站

Rank: 2

积分
162 
威望
162  
包包
1746  
藤椅
发表于 2015-6-25 11:27 |只看该作者
呵呵 大家好奇嘛 来观看下~~~~  

Rank: 2

积分
162 
威望
162  
包包
1724  
板凳
发表于 2015-8-11 17:35 |只看该作者
干细胞之家微信公众号
转基因动物

Rank: 2

积分
162 
威望
162  
包包
1746  
报纸
发表于 2015-8-21 10:44 |只看该作者
观看中  

Rank: 2

积分
97 
威望
97  
包包
1738  
地板
发表于 2015-8-25 18:49 |只看该作者
今天临床的资料更新很多呀

Rank: 2

积分
161 
威望
161  
包包
1862  
7
发表于 2015-8-30 12:15 |只看该作者
顶你一下,好贴要顶!  

Rank: 2

积分
69 
威望
69  
包包
1788  
8
发表于 2015-9-2 19:34 |只看该作者
干细胞行业  

Rank: 2

积分
136 
威望
136  
包包
1877  
9
发表于 2015-9-6 22:18 |只看该作者
干细胞抗衰老  

Rank: 2

积分
75 
威望
75  
包包
2193  
10
发表于 2015-9-11 00:48 |只看该作者
你加油吧  
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-5-5 23:27

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.