Take a look at the Recent articles

Evaluation of the impact of treatment delays and dose reductions in human ovarian cancer orthotopic mouse models

Scott Mosley

Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainsville, FL, USA

Jing-Hong Chen

Shanghai Jiao Tong University School of Medicine, Shanghai, China

Anjali Gaikwad

Department of Obstetrics, Gynecology and Reproductive Sciences, University of Texas Health Science Center Medical School at Houston; Houston, TX, USA

Elizabeth K. Nugent

Department of Obstetrics, Gynecology and Reproductive Sciences, University of Texas Health Science Center Medical School at Houston; Houston, TX, USA

Judith A. Smith

Department of Obstetrics, Gynecology and Reproductive Sciences, University of Texas Health Science Center Medical School at Houston; Houston, TX, USA

E-mail : Judith.Ann.Smith@uth.tmc.edu

DOI: 10.15761/ICST.1000155

Article
Article Info
Author Info
Figures & Data

Abstract

The primary objective of this study was to quantify the impact of treatment delay and dose reductions in an in vivo orthotopic ovarian cancer model.  In addition, this study evaluated the role of growth factors in achieving “full dose on time”. HeyA-8 and SKOV3.ip1ovarian cancer orthotopic mouse models were used determine the effect of paclitaxel/carboplatin treatment delays or dose reductions on tumor response and overall survival.  The addition of growth factor support with pegfilgrastim and darbepoetin was also evaluated in treatment control arm. The results indicated that full dose of paclitaxel (20 mg/kg) and carboplatin (50 mg/kg) delivered on time (every 21 days) achieved better tumor response in both aggressive (HeyA-8) and metastatic (SKOV3.ip1) human ovarian tumors compared to treatment delay or dose reduction arms.   In the HeyA-8 orthotopic model treatment delay (P<0.05) resulted in a significant reduction in overall survival compared to on time, full dose treatment control arm and the dose reduction decreased overall survival but was not statistically significant.  However, in the SKOV3.ip1orthotopic model a treatment delay (P<0.01) or dose reduction (P<0.05) both resulted in a significant reduction in overall survival compared to on time, full dose treatment control arm.  In both orthotopic models, the addition of pegfilgrastim (1000 µg/kg) and darbepoetin (10 µg/kg) delayed the onset of ascites formation and was associated with an improved overall survival rate.  Treatment delays and dose reductions had a negative impact on tumor response and overall survival compare with on-time, full dose treatment controls. In addition, use of growth factor agents improved overall survival and tumor responses.

Key words

dose reductions, treatment delays, chemotherapy, ovarian cancer

Introduction

Ovarian cancer is the most lethal gynecological cancer and the fifth most common cause of malignancy in women [1].  Ovarian cancer is difficult to detect and diagnose early, therefore patients often initially present with advanced disease and poor prognosis.   The current standard of care includes primary tumor debulking surgery followed by cytotoxic chemotherapy with 70% of patients achieving a complete initial clinical response.   The current standard of care for primary chemotherapy is six cycles of combination of taxanes agent plus a platinum agent given once every three weeks [2].  Although multiple studies have evaluated different dosing parameters and routes of administration to reduce toxicity and optimize efficacy, often clinical practice requires dose reductions or treatment delays to complete six cycles of therapy [3,4].

It is well established that granulocyte-colony stimulating factor (G-CSF) minimizes the magnitude and duration of neutropenia and improves overall treatment outcomes primarily by decreasing the risk of infection, hospitalization, and potentially fatal complications from sepsis [5-9].   In addition to increasing the risk of developing febrile neutropenia, prolonged neutropenia often leads to delays in treatment and dose reductions of planned chemotherapy [7,8].  Anemia is another common development during chemotherapy that is responsible for dose reductions and treatment delays; and prevention of anemia has been suggested to improve treatment outcomes both efficacy and quality of life.  Intratumoral hypoxia is associated with promoting resistance to chemotherapy and/or radiotherapy, genetic instability, angiogenesis, and inhibition of apoptosis [9-12].  New and improved growth factor support with agents such as pegfilgrastim and darbepoetin alfa have the potential to improve the management of chemotherapy-induced neutropenia and anemia, respectively [13].

There is minimal information about the impact of treatment delays or dose reductions on treatment responses and overall outcomes in cancer patients. The primary objective of this study was to quantify the impact of dose reductions and treatment delays in an in vivo orthotopic ovarian cancer model.   In addition, the role of growth factors to improve the ability of patients to receive “full dose on time” was evaluated.

Materials and methods

Cell culture

HeyA-8 human ovarian cancer cells obtained from American Type Culture Collection (Rockville, MD) were cultured in RPMI 1640 supplemented (GIBCO) with 10% fetal bovine serum (Invitrogen), SKOV3.ip1 human ovarian cancer cells were generously provided by Dr. Josh Fidler, The University of Texas, MD Anderson Cancer Center (UTMDACC) were cultured in Eagle’s Minimum Essential Medium (MEM) with Earle’s basic salt solution (BSS) and non-essential amino acids (NEAA), 1 mmol/L sodium pyruvate, 2X MEM vitamin solution with 10% fetal bovine serum and 1.5 mmol/L L-glutamine. Cell lines used for this study were less than 10 passages to ensure the platinum resistance or sensitivity, as well as preventing major changes in cell line characteristics.

Drugs

Paclitaxel 6 mg/mL solution (Eli Lilly, Inc Indianapolis, IN) and carboplatin 10 mg/mL solution (Eli Lilly, Inc Indianapolis, IN) were purchased from the UTMDACC pharmacy department. All drugs were diluted in fresh 0.9% sodium chloride immediately before each drug administration. Growth factor agents including pegfilgrastim 10 mg/mL and darbepoetin 10 mg/mL were generously provided from Amgen Oncology Company (Thousand. Oaks, CA).

HeyA-8 and SKOV3.ip1ovarian cancer orthotopic mouse models

The protocol was reviewed and approved by the institutional animal care and utilization committee (IACUC) prior to initiating any animal work.  All mice were handled according to the Guide for the Care and Use of Laboratory Animals.  HeyA-8 or SKOV3.ip1 cells were grown to confluence and harvested by trypsinization with 0.25 mg/mL trypsin/EDTA and suspended in PBS before inoculation into mice.  HeyA-8 cells (3 × 106) and SKOV3.ip1 cells (9 × 106) were collected, counted and dispersed in a volume of 200 µL of PBS or serum-free cell culture medium and were injected into the peritoneal cavities of six-week-old female nude mice 

Two intraperitoneal (IP) ovarian cancer orthotopic models were utilized including the HeyA-8 orthotopic model to represent chemosenstive, aggressive ovarian cancer or SKOV3.ip1 to represent multi-drug resistant metastatic ovarian cancer, to evaluate the dose and timing of administration of taxane/platinum combination regimen.   There were a total of six groups including: untreated, vehicle alone, treatment control A paclitaxel/carboplatin alone once every three weeks, and treatment control B paclitaxel/carboplatin + GCSF once every three weeks, experimental treatment delay group and experimental dose reduction group  for each orthotopic model with ten mice in each group (Figure 1).  A total of 90 mice per each cell line and 180 mice were required to complete the study.

Figure 1. Study Treatment flow chart. 90 mice per experiment two cell lines =* 180 mice

Treatment

Ten days after tumor inoculation, mice were randomized to receive administration of IV injection of normal saline, or paclitaxel 20 mg/kg IV and carboplatin 50 mg/kg IV alone, or paclitaxel 20 mg/kg IV and carboplatin 50 mg/kg IV on day one then pegfilgrastim 1000 µg/kg subcutaneous (subQ) and darbepoetin 10 µg/kg, subQ on day two.  Doses selected for this study were based on previous animal studies and are comparable to respective clinical doses tolerated in humans [14].

Each treatment group initiated chemotherapy with paclitaxel 20 mg/kg IV and carboplatin 50 mg/kg IV on day one of the each 21-day cycle with a total of three planned cycles.  The control treatment groups continued to receive full dose paclitaxel 20 mg/kg IV  and carboplatin 50 mg/kg IV on day one followed by pegfilgrastim 1000 µg/kg subQ + darbepoetin 10 µg/kg subQ on day two, every 21 days, for three cycles.  In the treatment delay experiment arm, there were empiric treatment delays in two treatment delay groups occurring on either cycle 2 or cycle 3, respectively (Figure 1).   In the dose reduction experimental arm there were empiric dose reductions of 25% in two of the dose reduction groups occurring on either cycle 2 or cycle 3, respectively (Figure 1).

Tumor response determination

As surrogates to reflect both increasing ascites accumulation and tumor burden the body weight and abdominal circumference were quantified three times a week as average measurement for each parameter.  Abdominal circumference measurements were obtained with a special device (restraining tube with fixed soft ruler) at the body’s widest mid-point marked and monitored throughout the experimental period while the animal was held in a prone position.  In addition, mice were monitored three times a week for evidence of signs/symptoms of moriboundity (weight loss, anorexia, hunching, listlessness, extensively enlarged abdominal cavity, lethargy, etc.). Paracentesis was performed to remove fluid up to 1 mL as appropriate to relieve abdominal bloating and fluid volume was measured up to three times for each animal before sacrificing.  At the end of the experiment, all remaining mice were sacrificed humanely via CO2 inhalation followed by cervical dislocation.

Data analysis

Descriptive statistics were completed to evaluate delay in tumor growth and/or tumor regression and contrasted with respective to toxicity profiles.  T-test analysis was used in the data analysis to evaluate and compare the activity and toxicity, respectively, for each experimental arm compared to the treatment control arms with and without growth factors.

Results

A summary of the impact of dose reductions and treatment delays on the overall survival rate of the HeyA-8 and SKOV3.ip1 tumor bearing mice is reported in Figure 2 and Table 1.

Figure 2. Summary figures of the overall survival. The survival curves of the six groups illustrating the survival of mice non-treated and different treatment arms for the HeyA-8 ovarian cancer orthotopic model (2A) and SKOV3.ip1ovarian cancer orthotopic model (2B). Survival rate of paclitaxel (20 mg/kg) + carboplatin (50 mg/kg) +/- growth factor support groups showed a significant improvement in the survival rate.

Cell type

Treatment Group

Ave Survived days ± SE

Bodyweight change ± SE

HeyA-8

Non-treatment control

37.5 ± 1.1

5.5 ± 0.6

Vehicle            control

36.5 ± 2

4.2 ± 0.9

Paclitaxel/Carboplatin treatment control

44.5 ± 4.9

2.4 ± 0.5***#

Paclitaxel/Carboplatin +GF control

55 ± 5.8** ##

1.6 ± 0.3*** #

Treatment delay (H1A)                       

30.5 ± 4.2^

2.6 ± 0.5**

Dose reduction (H2A)

44 ± 3.8

1.3 ± 0.3***#

SKOV3-IP1

Non-treatment control

38.5  ± 0.8

4.5  ± 0.6

Vehicle            control

38 ± 1.3

4.6 ± 0.7

Paclitaxel/Carboplatin treatment control

46.5 ± 1.8***###

2.9 ± 0.9

Paclitaxel/Carboplatin +GF control

44.5 ± 3.2* #

1.7 ± 0.7**##

Treatment delay (S1A)            

38 ± 2.4^^

4 ± 1

Dose reduction (S2A)

39 ± 2.8^

4.2 ± 0.5

Table 1. Overall Survival and living status change summary for HeyA-8 and SKOV3.ip1mice 

In the HeyA-8 non-treatment and vehicle control group, all mice died before day 40. The mean survival time of the HeyA-8 vehicle control group was 36.5 ± 2 days after tumor implantation. In the HeyA-8 paclitaxel/carboplatin treatment control group, however, two animal deaths occurred during the study and the remaining animals were still alive on day 40. The mean overall survival time in the HeyA-8 paclitaxel/carboplatin treatment group was 44.5 ± 4.9 days (Figure 2a).

In the HeyA-8 treatment delay group, four mice died during the study.  The mean overall survival was 30.5 ± 4.2 days which was significantly reduced compared to the treatment control groups (P<0.05)   (Table 1). For the HeyA-8 dose reduction group, the mean overall survival was 44 ± 3.8 days which was not statistically significant compared to the HeyA-8 full dose treatment control group.

Among the treatment groups, paclitaxel/carboplatin followed by growth factor (GF) agents had the longest median survival and the most surviving (65 days, 1 surviving mouse).  In the HeyA-8 paclitaxel/carboplatin +GF control group, the two animal deaths occurred during the study and the remaining animals were still alive after day 50.  The mean overall survival time was 55 ± 5.8 days which was statistically significant compared to the HeyA-8 untreated controls (P<0.01) (Figure 1a).   Paclitaxel/carboplatin +GF was improved but did not statistically differ compared to full dose, on time treatment control group (p=0.09). However, Paclitaxel/carboplatin +GF was superior to both the treatment delay (p<0.001) and dose reduction group (p=0.06).

Similar findings were observed in the multi-drug resistant SKOV3.ip1 orthotopic model study.  In SKOV3.ip1 arm nine of the non-treatment or vehicle control mice died before the day 40 and only one mouse survived until day 45. The mean overall survival time of the SKOV3.ip1vehicle group was 38 ± 1.33 days after tumor inoculation.   In the SKOV3.ip1full dose, on time paclitaxel/carboplatin treatment control group, seven animal deaths prior to day 50 post inoculation. The mean overall survival time in the SKOV3.ip1treatment control group was 46.5 ± 1.8 days (P<0.001).  In the SKOV3.ip1paclitaxel/carboplatin +GF group there were no deaths prior to day 55 post inoculation.  The mean overall survival for the SKOV3.ip1paclitaxel/carboplatin +GF group was 44.5 ± 3.2 days which was not statistically different than the SKOV3.ip1full dose, on time paclitaxel/carboplatin treatment control group (Figure 2b).  

In the SKOV3.ip1treatment delay group all mice died prior to day 45 of the study.  The mean overall survival was 38.0 ± 2.4 days which was significantly reduced compared to the full dose, on time paclitaxel/carboplatin treatment control group (p<0.01).  The SKOV3.ip1dose reduction group had a mean overall survival of 39 ± 2.8 days which was also significantly reduced compared to the full dose, on time paclitaxel/carboplatin treatment control group (P<0.05).  

Evaluation of ascites formation

At postmortem examination, tumors were found on the surface of the peritoneum, uterus on the diaphragm, in the liver, and on the intestines in both control and treated groups.   Ascites volumes were quantified during each experiment and postmortem examination. Groups were compared and the results are depicted in Table 2. The ascites onset in the HeyA-8 non-treatment and vehicle control groups was very early with the mean onset of ascites formation of 2.9 ± 0.8 and 33.1 ± 0.8 days, respectively. However, in the HeyA-8 paclitaxel/carboplatin + GF control group, a significant delay in the ascites formation was observed with a mean onset time of 53.1 ± 5 days (P<0.01).  A similar improvement was observed in the SKOV3.ip1 paclitaxel/carboplatin + GF control group having delayed onset of ascites formation compared with non-treatment groups (p<0.05).   No difference in the onset of ascites formation was observed between the non-treatment and treatment delay or dose reduction groups in either the HeyA-8 or SKOV3.ip1orthotopic models in this study.

Cell type

Treatment

Ave days onset of ascites ± SE

Ave ascites volume ± SE

Ave waistline changes ± SE

HeyA-8

non-treatment control

32.9 ± 0.8

0.9 ± 0.2

1.5 ± 0.2

Vehicle            control group

33.1 ± 0.8

1.1 ± 0.4

1.3 ± 0.2

Paclitaxel/Carboplatin treatment control

42.5 ± 5.6

0.45 ± 0.02 *

0.4 ± 0.2***###

Paclitaxel/Carboplatin +GF control                

53.1 ± 5 **##

0.7 ± 0.2

0.3 ± 0.1***###

Treatment delay (H1A)             

36 ± 2.3

0.8 ± 0.1

1 ± 0.2^

Dose reduction (H2A)

31.3 ± 1.5

0.7 ± 0.1

0.5 ± 0.2*

SKOV3-IP1

non-treatment control

31.5  ± 1.5      

0.8  ± 0.1

1.6 ± 0.1

Vehicle            control grou    

31.7 ± 1.1

0.7 ± 0.2

1.3 ± 0.2

Paclitaxel/Carboplatin treatment control

40 ± 1.9*

0.4 ± 0.1

0.4 ± 0.1***###

Paclitaxel/Carboplatin treatment+GF control           

40 ± 2*

0.5 ± 0.04 #

0.4 ±  0.1***###

Treatment delay (S1A)

33.3 ±  0.8

1.1 ± 0.4^

1.3 ± 0.2^

Dose reduction (S2A)

37 ± 2.1

0.7 ± 0.1

1.1 ± 0.3^

Table 2. The ascites formation analysis table for HeyA-8 and SKOV3-IP1 mice

Note: Symbol * is compare to Un-treatment group.  Symbol * is compare to Vehicle group.

         Symbol ^ is compare to Carboplatin+ Paclitaxel group.

 Note: Symbol *  compared to non-treatment control group. Symbol # compared to vehicle control group.         

Symbol ^  compared to paclitaxel/carboplatin treatment control group. *For statistically significance, one symbol indicated P<0.05, two symbols indicated P<0.01 and three symbols indicated P<0.001

Discussion

There is minimal information about the impact of treatment delays or dose reductions on treatment responses and overall outcomes.   There have been multiple retrospective clinical studies that have clearly demonstrated dose reductions are associated with decrease response rate and overall survival [15-19]. These studies have all evaluated the impact of dose reductions retrospectively and there are always concerns of bias or other variables that may confound results commonly associated with the retrospective study approach.   The impact of treatment delays has not been formally evaluated in the clinical setting due to feasibility of creating a homogenous patient population for an un-bias data analysis.  However, there is enough retrospective data to support that it would be unethical to attempt to conduct a prospective study to evaluate the impact of chemotherapy treatment delays and dose reductions in human ovarian cancer patients.  Hence, the use of human ovarian cancer orthotopic mouse model was ideal to prospectively evaluate the impact of both chemotherapy treatment delays and dose reductions in a controlled setting in a homogenous population.  This minimizes impact of outside confounders such as co-morbidities, concomitant medications, previous treatments, differing histology or tumor burden.

In both the HeyA-8 orthotopic model representing the aggressive chemosensitive newly diagnosed ovarian cancer and the SKOV3.ip1orthotopic model representing the multi-drug resistant or recurrent ovarian cancer, a decrease in overall survival and ascites formation was observed compared to the full dose on time paclitaxel/carboplatin treatment control group.  In addition, the use of growth factors, pegfilgrastim and darbepoetin, significantly improved overall survival and delayed the onset of ascites formation.  We hypothesis the addition of growth factors improved the ability of mice to tolerate full dose, on-time chemotherapy which resulted in an improvement in overall survival and tumor response.

This is a prospective study to quantify the impact of dose reductions and treatment delays in an in vivo human ovarian cancer model.   This study confirms what in the clinical setting intuitively has been suggested; that both treatment delays and dose reductions have a negative effect on treatment response.  This new data can be used to help define the importance of full dose and on time dose of the taxane/platinum agents used in treatment of newly diagnosed and recurrent ovarian cancer.  Furthermore, this study demonstrated the benefit of prospective use of growth factors, specifically pegfilgrastim, in combination with the taxane/platinum regimen improved tumor response and ultimately overall survival. 

Acknowledgement

Study was sponsored by unrestricted research grant from Amgen Oncology, INC.

2021 Copyright OAT. All rights reserv

References

  1. Guttman O, Baranovski BM, Schuster R, Kaner Z, Freixo-Lima GS, et al. (2015) Acute-phase protein α1-anti-trypsin: diverting injurious innate and adaptive immune responses from non-authentic threats. Clin Exp Immunol 179: 161-172. [Crossref]
  2. Gottlieb PA, Alkanani AK, Michels AW, Lewis EC, Shapiro L, et al. (2014) α1-Antitrypsin therapy downregulates toll-like receptor-induced IL-1beta responses in monocytes and myeloid dendritic cells and may improve islet function in recently diagnosed patients with type 1 diabetes. J Clin Endocrinol Metab 99: E1418-1426. [Crossref]
  3. Marcondes AM, Karoopongse E, Lesnikova M, Margineantu D, Welte T, et al. (2014) α-1-Antitrypsin (AAT)-modified donor cells suppress GVHD but enhance the GVL effect: a role for mitochondrial bioenergetics. Blood 124: 2881-2891. [Crossref]
  4. Hagen LE, Schechter T, Luk Y, Brodovitch A, Gassas A, et al. (2011) High alpha-1 antitrypsin clearance predicts severity of gut graft-versus-host disease (GVHD) in children. Pediatr Transplant 15: 659-663. [Crossref]
  5. Tawara I, Sun Y, Leewis EC, Toubai T, Evers R, et al. (2012) Alpha-1-antitrypsin monotherapy reduces graft-versus-host disease after experimental allogeneic bone marrow transplantation. Proc Natl Acad Sci U S A 109: 564-569. [Crossref]
  6. Subramanian S, Shahaf G, Ozeri E, Miller LM, Vandenbark AA, et al. (2011) Sustained expression of circulating human alpha-1 antitrypsin reduces inflammation, increases CD4+FoxP3+ Treg cell population and prevents signs of experimental autoimmune encephalomyelitis in mice. Metab Brain Dis 26: 107-113. [Crossref]
  7. Collins CB, Aherne CM, Ehrentraut SF, Gerich ME, McNamee EN, et al. (2013) Alpha-1-antitrypsin therapy ameliorates acute colitis and chronic murine ileitis. Inflamm Bowel Dis 19: 1964-1973. [Crossref]
  8. Breit SN, Wakefield D, Robinson JP, Luckhurst E, Clark P, et al. (1985) The role of alpha 1-antitrypsin deficiency in the pathogenesis of immune disorders. Clin Immunol Immunopathol 35: 363-380.[Crossref]
  9. Zhang B, Lu Y, Campbell-Thompson M, Spencer T, Wasserfall C, et al. (2007) Alpha1-antitrypsin protects beta-cells from apoptosis. Diabetes 56: 1316-1323. [Crossref]
  10. Kataoka H, Uchino H, Iwamura T, Seiki M, Nabeshima K, et al. (1999) Enhanced tumor growth and invasiveness in vivo by a carboxyl-terminal fragment of alpha1-proteinase inhibitor generated by matrix metalloproteinases: a possible modulatory role in natural killer cytotoxicity. Am J Pathol 154: 457-468. [Crossref]
  11. Okada Y, Nakanishi I (1989) Activation of matrix metalloproteinase 3 (stromelysin) and matrix metalloproteinase 2 ('gelatinase') by human neutrophil elastase and cathepsin G. FEBS Lett 249: 353-356. [Crossref]
  12. Carlson JA, Rogers BB, Sifers RN, Hawkins HK, Finegold MJ, et al. (1988) Multiple tissues express alpha 1-antitrypsin in transgenic mice and man. J Clin Invest 82: 26-36. [Crossref]
  13. Kaner Z, Ochayon DE, Shahaf G, Baranovski BM, Bahar N, et al. (2015) Acute phase protein α1-antitrypsin reduces the bacterial burden in mice by selective modulation of innate cell responses. J Infect Dis 211: 1489-1498. [Crossref]
  14. Brantly M, Nukiwa T, Crystal RG (1988) Molecular basis of alpha-1-antitrypsin deficiency. Am J Med 84: 13-31. [Crossref]
  15. Lewis EC (2012) Expanding the clinical indications for α(1)-antitrypsin therapy. Mol Med 18: 957-970. [Crossref]
  16. Mizrahi M, Cal P, Rosenthal M, Ochayon D, Shahaf G,et al. (2013) Human alpha1-antitrypsin modifies B-lymphocyte responses during allograft transplantation. Immunology 140: 362-373. [Crossref]
  17. Churg A, Wang X, Wang RD, Meixner SC, Pryzdial EL, et al. (2007) Alpha1-antitrypsin suppresses TNF-alpha and MMP-12 production by cigarette smoke-stimulated macrophages. Am J Resp Cell Mol Biol 37:144-151. [Crossref]
  18. Ozeri E, Mizrahi M, Shahaf G, Lewis EC (2012) α-1 antitrypsin promotes semimature, IL-10-producing and readily migrating tolerogenic dendritic cells. J Immunol 189: 146-153. [Crossref]
  19. Guttman O, Yossef R, Freixo-Lima G, Rider P, Porgador A, et al. (2014) α1-Antitrypsin modifies general NK cell interactions with dendritic cells and specific interactions with islet β-cells in favor of protection from autoimmune diabetes. Immunology. [Crossref]
  20. Lewis EC, Mizrahi M, Toledano M, DeFeliceN, Wright JL, et al. (2008) alpha1-Antitrypsin monotherapy induces immune tolerance during islet allograft transplantation in mice. Proc Natl Acad Sci U S A 105: 16236-16241. [Crossref]
  21. Fairbanks KD, Tavill AS (2008) Liver disease in alpha 1-antitrypsin deficiency: a review. Am J Gastroenterol 103: 2136-2141. [Crossref]
  22. Eriksson S, Carlson J, Velez R (1986) Risk of cirrhosis and primary liver cancer in alpha 1-antitrypsin deficiency. N Engl J Med 314: 736-739. [Crossref]
  23. Rudnick DA, Perlmutter DH (2005) Alpha-1-antitrypsin deficiency: a new paradigm for hepatocellular carcinoma in genetic liver disease. Hepatology 42: 514-521. [Crossref]
  24. Yang P, Wentzlaff KA, Katzmann JA, Marks RS, Allen MS,et al. (1999) Alpha1-antitrypsin deficiency allele carriers among lung cancer patients. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology 8: 461-465.
  25. Yang P, Sun Z, Krowka MJ, Aubry MC, Bamlet WR, et al. (2008) Alpha1-antitrypsin deficiency carriers, tobacco smoke, chronic obstructive pulmonary disease, and lung cancer risk. Arch Intern Med 168: 1097-1103. [Crossref]
  26. Yang P, Cunningham JM, Halling KC, Lesnick TG, Burgart LJ, et al. (2000) Higher risk of mismatch repair-deficient colorectal cancer in alpha(1)-antitrypsin deficiency carriers and cigarette smokers. Mol Genet Metab 71: 639-645. [Crossref]
  27. Sun Z, Yang P (2004) Role of imbalance between neutrophil elastase and alpha 1-antitrypsin in cancer development and progression. Lancet Oncol 5: 182-190. [Crossref]
  28. Sparos L, Tountas Y, Chapuis-Cellier C, Theodoropoulos G, Trichopoulos D (1984) Alpha 1-antitrypsin levels and phenotypes and hepatitis B serology in liver cancer. Br J Cancer 49: 567-570. [Crossref]
  29. Harris CC, Cohen MH, Connor R, Primack A, Saccomanno G, et al. (1976) Serum alpha1-antitrypsin in patients with lung cancer or abnormal sputum cytology. Cancer 38: 1655-1657. [Crossref]
  30. Harris CC, Primack A, Cohen MH (1974) Elevated alpha1-Antitrypsin serum levels in lung cancer patients. Cancer 34: 280-281. [Crossref]
  31. Karashima S, Kataoka H, Itoh H, Maruyama R, Koono M (1990) Prognostic significance of alpha-1-antitrypsin in early stage of colorectal carcinomas. Int J Cancer 45: 244-250. [Crossref]
  32. Lopez-Arias E, et al. (2012) Alpha 1-antitrypsin: a novel tumor-associated antigen identified in patients with early-stage breast cancer. Electrophoresis 33: 2130-2137.
  33. Tountas Y, Sparos L, Theodoropoulos C, Trichopoulos D (1985) Alpha 1-antitrypsin and cancer of the pancreas. Digestion 31: 37-40. [Crossref]
  34. Urquidi V, Goodison S, Ross S, Chang M, Dai Y, et al. (2012) Diagnostic potential of urinary alpha1-antitrypsin and apolipoprotein E in the detection of bladder cancer. J Urol 188: 2377-2383. [Crossref]
  35. Rostenberg I, Guízar-Vázquez J, Peñaloza R (1978) Altered carbohydrate content of alpha1-antitrypsin in patients with cancer. J Natl Cancer Inst 61: 961-965. [Crossref]
  36. Thompson S, Guthrie D, Turner GA (1988) Fucosylated forms of alpha-1-antitrypsin that predict unresponsiveness to chemotherapy in ovarian cancer. Br J Cancer 58: 589-593. [Crossref]
  37. Comunale MA, Rodemich-Betesh L, Hafner J, Wang M, Norton P, et al. (2010) Linkage specific fucosylation of alpha-1-antitrypsin in liver cirrhosis and cancer patients: implications for a biomarker of hepatocellular carcinoma. PloS One 5: e12419. [Crossref]
  38. Huang H, Campbell SC, Nelius T, Bedford DF, Veliceasa D, et al. (2004) Alpha1-antitrypsin inhibits angiogenesis and tumor growth. Int J Cancer 112: 1042-1048. [Crossref]
  39. Zelvyte I, Stevens T, Westin U, Janciauskiene S (2004) α1-antitrypsin and its C-terminal fragment attenuate effects of degranulated neutrophil-conditioned medium on lung cancer HCC cells, in vitro. Cancer Cell Int 4: 7. [Crossref]
  40. Finlay TH, Tamir S, Kadner SS, Cruz MR, Yavelow J, et al. (1993) alpha 1-Antitrypsin- and anchorage-independent growth of MCF-7 breast cancer cells. Endocrinology 133: 996-1002. [Crossref]
  41. Tamir S, Kadner SS, Katz J, Finlay TH (1990) Regulation of antitrypsin and antichymotrypsin synthesis by MCF-7 breast cancer cell sublines. Endocrinology 127: 1319-1328. [Crossref]
  42. Yavelow J, Tuccillo A, Kadner SS, Katz J, Finlay TH (1997) Alpha 1-antitrypsin blocks the release of transforming growth factor-alpha from MCF-7 human breast cancer cells. J Clin Endocrinol Metab 82: 745-752. [Crossref]
  43. Solinas G, Germano G, Mantovani A, Allavena P (2009) Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J Leukoc Biol 86: 1065-1073. [Crossref]
  44. Principe DR, Doll JA, Bauer J, Jung B, Munshi HG, et al. (2014) TGF-β: duality of function between tumor prevention and carcinogenesis. J Natl Cancer Inst 106: djt369. [Crossref]
  45. Laouar Y, Sutterwala FS, Gorelik L, Flavell RA (2005) Transforming growth factor-beta controls T helper type 1 cell development through regulation of natural killer cell interferon-gamma. Nat Immunol 6: 600-607. [Crossref]
  46. Yu Q, Stamenkovic I (2000) Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis. Genes Dev 14: 163-176. [Crossref]
  47. Csernok E, Szymkowiak CH, Mistry N, Daha MR, Gross WL,et al. (1996) Transforming growth factor-beta (TGF-beta) expression and interaction with proteinase 3 (PR3) in anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis. Clin Exp Immunol 105: 104-111. [Crossref]
  48. Serban KA,Petrusca DN, Lockett AD, Justice MJ, Saint L, et al. (2015) Alpha-1 Antitrypsin (AAT) Enhances Alveolar Macrophages Scavenging During Conditions Simulating COPD Exacerbation. B110. COPD GALORE: NEW INSIGHTS INTO BRONCHITIS AND EMPHYSEMA DEVELOPMENT AND TREATMENT. American Thoracic Society International Conference Abstracts, (American Thoracic Society): A3882-A3882.
  49. Mencin A, Seki E, Osawa Y, Kodama Y, De Minicis S, et al. (2007) Alpha-1 antitrypsin Z protein (PiZ) increases hepatic fibrosis in a murine model of cholestasis. Hepatology 46: 1443-1452. [Crossref]
  50. Subramanian S, Shahaf G, Ozeri E, Miller LM, Vandenbark AA, et al. (2011) Sustained expression of circulating human alpha-1 antitrypsin reduces inflammation, increases CD4+FoxP3+ Treg cell population and prevents signs of experimental autoimmune encephalomyelitis in mice. Metab Brain Dis 26: 107-113. [Crossref]
  51. Tawara I, Sun Y, Lewis EC, Toubai T, Evers R, et al. (2012) Alpha-1-antitrypsin monotherapy reduces graft-versus-host disease after experimental allogeneic bone marrow transplantation. Proc Natl Acad Sci U S A 109: 564-569. [Crossref]
  52. Hodge DR, Hurt EM, Farrar WL (2005) The role of IL-6 and STAT3 in inflammation and cancer. Eur J Cancer 41: 2502-2512. [Crossref]
  53. Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, et al. (2009) IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell 15: 103-113. [Crossref]
  54. Mantovani A, Allavena P, Sica A, Balkwill F (2008) Cancer-related inflammation. Nature 454: 436-444. [Crossref]
  55. Grivennikov SI, Karin M (2011) Inflammatory cytokines in cancer: tumour necrosis factor and interleukin 6 take the stage. Ann Rheum Dis 70: i104-108. [Crossref]
  56. Locasale JW, Zeskind B (2011) IL-6 and ovarian cancer--letter. Clin Cancer Res 17: 7837. [Crossref]
  57. Neurath MF, Finotto S (2011) IL-6 signaling in autoimmunity, chronic inflammation and inflammation-associated cancer. Cytokine Growth Factor Rev 22: 83-89. [Crossref]
  58. Petrache I, Fijalkowska I, Zhen L, Medler TR, Brown E, et al. (2006) A novel antiapoptotic role for alpha1-antitrypsin in the prevention of pulmonary emphysema. Am J Respir Crit Care Med 173: 1222-1228. [Crossref]
  59. Raulet DH (2003) Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol 3: 781-790. [Crossref]
  60. Arnon TI, Achdout H, Lieberman N, Gazit R, Gonen-Gross T, et al. (2004) The mechanisms controlling the recognition of tumor- and virus-infected cells by NKp46. Blood 103: 664-672. [Crossref]
  61. Gur C, Enk J, Kassem SA, Suissa Y, Magenheim J, et al. (2011) Recognition and killing of human and murine pancreatic beta cells by the NK receptor NKp46. J Immunol 187: 3096-3103. [Crossref]
  62. Gur C, Porgador A, Elboim M, Gazit R, Mizrahi S, et al. (2010) The activating receptor NKp46 is essential for the development of type 1 diabetes. Nat Immunol 11: 121-128. [Crossref]
  63. Yossef R, Gur C, Shemesh A, Guttman O, Hadad U, et al. (2015) Targeting natural killer cell reactivity by employing antibody to NKp46: implications for type 1 diabetes. PLoS One 10: e0118936. [Crossref]
  64. Lewis EC, Shapiro L, Bowers OJ, Dinarello CA (2005) Alpha1-antitrypsin monotherapy prolongs islet allograft survival in mice. Proc Natl Acad Sci U S A 102: 12153-12158. [Crossref]
  65. Lu Y, Tang M, Wasserfall C, Kou Z, Campbell-Thompson M, et al. (2006) Alpha1-antitrypsin gene therapy modulates cellular immunity and efficiently prevents type 1 diabetes in nonobese diabetic mice. Hum Gene Ther 17: 625-634. [Crossref]
  66. Koulmanda M, Bhasin M, Hoffman L, Fan Z, Qipo A, et al. (2008) Curative and beta cell regenerative effects of alpha1-antitrypsin treatment in autoimmune diabetic NOD mice. Proc Natl Acad Sci U S A 105: 16242-16247. [Crossref]
  67. Ko K, Yamazaki S, Nakamura K, Nishioka T, Hirota K, et al. (2005) Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J Exp Med 202: 885-891. [Crossref]
  68. Chen ML, Pittet MJ, Gorelik L, Flavell RA, Weissleder R, et al. (2005) Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-beta signals in vivo. Proc Natl Acad Sci U S A 102: 419-424. [Crossref]
  69. Jarnicki AG, Lysaght J, Todryk S, Mills KH (2006) Suppression of antitumor immunity by IL-10 and TGF-beta-producing T cells infiltrating the growing tumor: influence of tumor environment on the induction of CD4+ and CD8+ regulatory T cells. J Immunol 177: 896-904. [Crossref]
  70. Balkwill F, Mantovani A (2001) Inflammation and cancer: back to Virchow? Lancet 357: 539-545. [Crossref]
  71. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A (2002) Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol 23: 549-555. [Crossref]
  72. Balkwill F, Charles KA, Mantovani A (2005) Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell 7: 211-217. [Crossref]
  73. Galdiero MR, Garlanda C, Jaillon S, Marone G, Mantovani A (2013) Tumor associated macrophages and neutrophils in tumor progression. J Cell Physiol 228: 1404-1412. [Crossref]
  74. Allavena P, Sica A, Solinas G, Porta C, Mantovani A (2008) The inflammatory micro-environment in tumor progression: the role of tumor-associated macrophages. Crit Rev Oncol Hematol 66: 1-9. [Crossref]
  75. Carmi Y, Dotan S, Rider P, Kaplanov I, White MR, et al. (2013) The role of IL-1β in the early tumor cell-induced angiogenic response. J Immunol 190: 3500-3509. [Crossref]
  76. Wu H, Xu JB, He YL, Peng JJ, Zhang XH, et al. (2012) Tumor-associated macrophages promote angiogenesis and lymphangiogenesis of gastric cancer. J Surg Oncol 106: 462-468. [Crossref]
  77. Chen P, Huang Y, Bong R, Ding Y, Song N, et al. (2011) Tumor-associated macrophages promote angiogenesis and melanoma growth via adrenomedullin in a paracrine and autocrine manner. Clin Cancer Res 17: 7230-7239. [Crossref]
  78. Sica A, Saccani A, Bottazzi B, Polentarutti N, Vecchi A, et al. (2000) Autocrine production of IL-10 mediates defective IL-12 production and NF-kappa B activation in tumor-associated macrophages. J Immunol 164: 762-767. [Crossref]
  79. Balkwill FR, Mantovani A (2012) Cancer-related inflammation: common themes and therapeutic opportunities. Semin Cancer Biol 22: 33-40. [Crossref]
  80. Mantovani A Biswas SK, Galdiero MR, Sica A, Locati M (2013) Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol 229: 176-185. [Crossref]
  81. Sica A, Mantovani A (2012) Macrophage plasticity and polarization: in vivo veritas. J Clin Invest 122: 787-795. [Crossref]
  82. Perlino E, Cortese R, Ciliberto G (1987) The human alpha 1-antitrypsin gene is transcribed from two different promoters in macrophages and hepatocytes. EMBO J 6: 2767-2771. [Crossref]
  83. van 'tWout EF, van Schadewijk A, Savage ND, Stolk J, Hiemstra PS (2012) α1-antitrypsin production by proinflammatory and antiinflammatory macrophages and dendritic cells. Am J Respir Cell Mol Biol 46: 607-613. [Crossref]
  84. Serban KA, Petrusca DN, Lockett AD, Justice MJ, Petrache I (2013) Alpha 1 Antitrypsin Effect On Alveolar Macrophages Engulfment Of Apoptotic And Phagocytic Targets. C68. AUTOPHAGY AND APOPTOSIS: NEW FEATURES OF CELL FATE. American Thoracic Society International Conference Abstracts, (American Thoracic Society): A4766-A4766.
  85. Abecassis A, Schuster R, Shahaf G, Ozeri E, Green R,et al. (2014) alpha1-antitrypsin increases interleukin-1 receptor antagonist production during pancreatic islet graft transplantation. Cell Mol Immunol 11: 377-386. [Crossref]
  86. Schouppe E, De Baetselier P, Van Ginderachter JA, Sarukhan A (2012) Instruction of myeloid cells by the tumor microenvironment: Open questions on the dynamics and plasticity of different tumor-associated myeloid cell populations. Oncoimmunology 1: 1135-1145. [Crossref]
  87. Hurwitz AA, Watkins SK (2012) Immune suppression in the tumor microenvironment: a role for dendritic cell-mediated tolerization of T cells. Cancer Immunol Immunother 61: 289-293.[Crossref]
  88. Fridman WH, Galon J, Pagès F, Tartour E, Sautès-Fridman C, et al. (2011) Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res 71: 5601-5605. [Crossref]
  89. Coussens LM, Werb Z (2002) Inflammation and cancer. Nature 420: 860-867. [Crossref]
  90. Smith HA, Kang Y (2013) The metastasis-promoting roles of tumor-associated immune cells. J Mol Med (Berl) 91: 411-429. [Crossref]
  91. Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, et al. (2004) NF-kappa B functions as a tumour promoter in inflammation-associated cancer. Nature 431: 461-466. [Crossref]
  92. Lockett AD, Kimani S, Ddungu G, Wrenger S, Tuder RM, et al. (2013) α₁-Antitrypsin modulates lung endothelial cell inflammatory responses to TNF-α. Am J Respir Cell Mol Biol 49: 143-150. [Crossref]
  93. Stetler-Stevenson WG, Aznavoorian S, Liotta LA (1993) Tumor cell interactions with the extracellular matrix during invasion and metastasis. Annu Rev Cell Biol 9: 541-573. [Crossref]
  94. Zheng H, Takahashi H, Murai Y, Cui Z, Nomoto K, et al. (2006) Expressions of MMP-2, MMP-9 and VEGF are closely linked to growth, invasion, metastasis and angiogenesis of gastric carcinoma. Anticancer Res 26: 3579-3583. [Crossref]
  95. John A, Tuszynski G (2001) The role of matrix metalloproteinases in tumor angiogenesis and tumor metastasis. Pathol Oncol Res 7: 14-23. [Crossref]
  96. Kleiner DE, Stetler-Stevenson WG (1999) Matrix metalloproteinases and metastasis. Cancer Chemother Pharmacol 43: S42-51. [Crossref]
  97. Joyce JA (2005) Therapeutic targeting of the tumor microenvironment. Cancer Cell 7: 513-520. [Crossref]
  98. Joyce JA, Pollard JW (2009) Microenvironmental regulation of metastasis. Nat Rev Cancer 9: 239-252. [Crossref]
  99. Sato N, Sutani A, Oya H, Yamaguchi T, Saito K, et al. (2007) Prognostic significance of neutrophil elastase inhibitor in patients with acute lung injury and interstitial pneumonia. Nihon Kokyuki Gakkaizasshi 45: 237-242. [Crossref]
  100. Inada M, Yamashita J, Nakano S, Ogawa M (1998) Complete inhibition of spontaneous pulmonary metastasis of human lung carcinoma cell line EBC-1 by a neutrophil elastase inhibitor (ONO-5046.Na). Anticancer Res 18: 885-890. [Crossref]
  101. Houghton AM (2013) Mechanistic links between COPD and lung cancer. Nat Rev Cancer 13: 233-245. [Crossref]
  102. DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, et al. (2009) CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 16: 91-102. [Crossref]
  103. Wang L, Yi T, Kortylewski M, Pardoll DM, Zeng D, et al. (2009) IL-17 can promote tumor growth through an IL-6-Stat3 signaling pathway. J Exp Med 206: 1457-1464. [Crossref]
  104. Mohamed MM, Cavallo-Medved D, Rudy D, Anbalagan A, Moin K, et al. (2010) Interleukin-6 increases expression and secretion of cathepsin B by breast tumor-associated monocytes. Cell Physiol Biochem 25: 315-324. [Crossref]
  105. Condeelis J, Pollard JW (2006) Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 124: 263-266. [Crossref]
  106. Camerer E, Qazi AA, Duong DN, Cornelissen I, Advincula R, et al. (2004) Platelets, protease-activated receptors, and fibrinogen in hematogenous metastasis. Blood 104: 397-401. [Crossref]
  107. Jurasz P, Alonso-Escolano D, Radomski MW (2004) Platelet--cancer interactions: mechanisms and pharmacology of tumour cell-induced platelet aggregation. Br J Pharmacol 143: 819-826. [Crossref]
  108. Nash GF, Turner LF, Scully MF, Kakkar AK (2002) Platelets and cancer. Lancet Oncol 3: 425-430. [Crossref]
  109. Gans H, Tan BH (1967) Alpha-1-antitrypsin, an inhibitor for thrombin and plasmin. Clin Chim Acta 17: 111-117. [Crossref]
  110. Cho HJ, Jung JI, Lim do Y, Kwon GT, Her S,et al. (2012) Bone marrow-derived, alternatively activated macrophages enhance solid tumor growth and lung metastasis of mammary carcinoma cells in a Balb/C mouse orthotopic model. Breast Cancer Res 14: R81. [Crossref]
  111. Gil-BernabéAM, Ferjancic S, Tlalka M, Zhao L, Allen PD, et al. (2012) Recruitment of monocytes/macrophages by tissue factor-mediated coagulation is essential for metastatic cell survival and premetastatic niche establishment in mice. Blood 119: 3164-3175. [Crossref]
  112. Yan HH, Pickup M, Pang Y, Gorska AE, Li Z, et al. (2010) Gr-1+CD11b+ myeloid cells tip the balance of immune protection to tumor promotion in the premetastatic lung. Cancer Res 70: 6139-6149.[Crossref]
  113. Lu X, Kang Y (2009) Chemokine (C-C motif) ligand 2 engages CCR2+ stromal cells of monocytic origin to promote breast cancer metastasis to lung and bone. J Biol Chem 284: 29087-29096. [Crossref]
  114. Janciauskiene S, Larsson S, Larsson P, Virtala R, Jansson L,et al. (2004) Inhibition of lipopolysaccharide-mediated human monocyte activation, in vitro, by alpha1-antitrypsin. Biochem Biophys Res Commun 321: 592-600. [Crossref]
  115. Bellacen K, Kalay N, Ozeri E, Shahaf G, Lewis EC (2013) Revascularization of pancreatic islet allografts is enhanced by α-1-antitrypsin under anti-inflammatory conditions. Cell Transplant 22: 2119-2133. [Crossref]
  116. Stockley RA, Turner AM (2014) α-1-Antitrypsin deficiency: clinical variability, assessment, and treatment. Trends Mol Med 20: 105-115. [Crossref]

Editorial Information

Editor-in-Chief

Masayoshi Yamaguchi
Emory University School of Medicine

Article Type

Research Article

Publication history

Received:October 10, 2015
Accepted: October 24, 2015
Published: October 27, 2015

Copyright

©2015Mosley S.This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Mosley S, Chen JH, Gaikwad A, Nugent EK, SmithJA(2015) Evaluation of the impact of treatment delays and dose reductions in human ovarian cancer orthotopic mouse models. Integr Cancer Sci Therap. 2: DOI: 10.15761/ICST.1000155

Corresponding author

Judith A. Smith

Department of Obstetrics, Gynecology and Reproductive Sciences, The University of Texas Health Sciences Center, 6431 Fannin Street, Rm 3.152, Houston, TX 77030, USA, Tel: 1-713-500-6408; Fax: 1-713-500-5474.

E-mail : Judith.Ann.Smith@uth.tmc.edu

Cell type

Treatment Group

Ave Survived days ± SE

Bodyweight change ± SE

HeyA-8

Non-treatment control

37.5 ± 1.1

5.5 ± 0.6

Vehicle            control

36.5 ± 2

4.2 ± 0.9

Paclitaxel/Carboplatin treatment control

44.5 ± 4.9

2.4 ± 0.5***#

Paclitaxel/Carboplatin +GF control

55 ± 5.8** ##

1.6 ± 0.3*** #

Treatment delay (H1A)                       

30.5 ± 4.2^

2.6 ± 0.5**

Dose reduction (H2A)

44 ± 3.8

1.3 ± 0.3***#

SKOV3-IP1

Non-treatment control

38.5  ± 0.8

4.5  ± 0.6

Vehicle            control

38 ± 1.3

4.6 ± 0.7

Paclitaxel/Carboplatin treatment control

46.5 ± 1.8***###

2.9 ± 0.9

Paclitaxel/Carboplatin +GF control

44.5 ± 3.2* #

1.7 ± 0.7**##

Treatment delay (S1A)            

38 ± 2.4^^

4 ± 1

Dose reduction (S2A)

39 ± 2.8^

4.2 ± 0.5

Table 1. Overall Survival and living status change summary for HeyA-8 and SKOV3.ip1mice 

Cell type

Treatment

Ave days onset of ascites ± SE

Ave ascites volume ± SE

Ave waistline changes ± SE

HeyA-8

non-treatment control

32.9 ± 0.8

0.9 ± 0.2

1.5 ± 0.2

Vehicle            control group

33.1 ± 0.8

1.1 ± 0.4

1.3 ± 0.2

Paclitaxel/Carboplatin treatment control

42.5 ± 5.6

0.45 ± 0.02 *

0.4 ± 0.2***###

Paclitaxel/Carboplatin +GF control                

53.1 ± 5 **##

0.7 ± 0.2

0.3 ± 0.1***###

Treatment delay (H1A)             

36 ± 2.3

0.8 ± 0.1

1 ± 0.2^

Dose reduction (H2A)

31.3 ± 1.5

0.7 ± 0.1

0.5 ± 0.2*

SKOV3-IP1

non-treatment control

31.5  ± 1.5      

0.8  ± 0.1

1.6 ± 0.1

Vehicle            control grou    

31.7 ± 1.1

0.7 ± 0.2

1.3 ± 0.2

Paclitaxel/Carboplatin treatment control

40 ± 1.9*

0.4 ± 0.1

0.4 ± 0.1***###

Paclitaxel/Carboplatin treatment+GF control           

40 ± 2*

0.5 ± 0.04 #

0.4 ±  0.1***###

Treatment delay (S1A)

33.3 ±  0.8

1.1 ± 0.4^

1.3 ± 0.2^

Dose reduction (S2A)

37 ± 2.1

0.7 ± 0.1

1.1 ± 0.3^

Table 2. The ascites formation analysis table for HeyA-8 and SKOV3-IP1 mice

Note: Symbol * is compare to Un-treatment group.  Symbol * is compare to Vehicle group.

Symbol ^ is compare to Carboplatin+ Paclitaxel group.

Note: Symbol *  compared to non-treatment control group. Symbol # compared to vehicle control group.         

Symbol ^  compared to paclitaxel/carboplatin treatment control group. *For statistically significance, one symbol indicated P<0.05, two symbols indicated P<0.01 and three symbols indicated P<0.001

Figure 1. Study Treatment flow chart. 90 mice per experiment two cell lines =* 180 mice

Figure 2. Summary figures of the overall survival. The survival curves of the six groups illustrating the survival of mice non-treated and different treatment arms for the HeyA-8 ovarian cancer orthotopic model (2A) and SKOV3.ip1ovarian cancer orthotopic model (2B). Survival rate of paclitaxel (20 mg/kg) + carboplatin (50 mg/kg) +/- growth factor support groups showed a significant improvement in the survival rate.