首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 46 毫秒
1.
There are a number of issues specific to breast cancer diagnosis in young women:
1 Breast cancer is uncommon in young women. It is associated with more-aggressive behavior and a worse prognosis. These clinical observations have recently been verified by histologic and biochemical analyses of these cancers.
2 Young women undergoing breast-conservation therapy can have a higher rate of local recurrence than older women, and this issue should be specifically addressed in preoperative counseling of these women.
3 Long life expectancy, issues of fertility, and risk of premature menopause resulting from cytotoxic chemotherapy are concerns unique to the young breast cancer patient.
4 Issues related to a new diagnosis of breast cancer associated with pregnancy bring up a multitude of issues in this vulnerable population.
5 Psychosocial issues of sexuality and self-image can warrant interventions in this population.

References

1 Surveillance Epidemiology and End Results Program, Surveillance, Epidemiology and End Results program public use CD-ROM, National Cancer Institute, Bethesda, MD (2000).
2 J.E.C. Kollias, I.O. Ellis, J.F. Robertson and R.W. Blamey, Early-onset breast cancer: histopathological and prognostic consideration, Br J Cancer 75 (1997), pp. 1318–1323. View Record in Scopus | Cited By in Scopus (98)
3 C. Shannon and I.E. Smith, Breast cancer in adolescents and young women, Eur J Cancer 39 (2003), pp. 2632–2642. Article | PDF (242 K) | View Record in Scopus | Cited By in Scopus (38)
4 R.A. Walker, E. Lees, M.B. Webb and S.J. Dearing, Breast carcinomas occurring in young women are different, Br J Cancer 74 (1996), pp. 1796–1800. View Record in Scopus | Cited By in Scopus (97)
5 D.P. Winchester, R.T. Osteen and H.R. Menck, The National Cancer Data Base report on breast cancer characteristics and outcome in relation to age, Cancer 78 (1996), pp. 1838–1843. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (83)
6 M. Chung, H.R. Chang, K.I. Bland and H.J. Wanebo, Younger women with breast cancer have a poorer prognosis than older women, Cancer 77 (1996), pp. 97–103. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (168)
7 H.O. Adami, B. Malker and L. Holmberg et al., The relationship between survival and age at diagnosis in breast cancer, N Engl J Med 315 (1986), pp. 559–563. View Record in Scopus | Cited By in Scopus (174)
8 Y.H. Wen, D.F. Roses and D.M. Axelrod et al., Breast carcinoma in women 30 years and younger: Abstract # 4005, Breast Cancer Res Treat 100 (2006), p. S264.
9 I. Matos, R. Dufloth and M. Alvarenga et al., p63, cytokeratin 5, and P-cadherin: three molecular markers to distinguish basal phenotype in breast cancer, Virchows Archive 447 (2005), pp. 688–694. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (62)
10 K.S. Albains, D.C. Allred and G.M. Clark, Breast cancer outcome and predictors of outcome: are there age differentials?, J Nat Cancer Inst Monogr 16 (1994), pp. 35–42.
11 K.I.J. Holli, Effect of age on the survival of breast cancer patients, Eur J Cancer 33 (1997), pp. 425–428. Abstract | PDF (498 K) | View Record in Scopus | Cited By in Scopus (38)
12 C. Smigal, A. Jemal and E. Ward et al., Trends in breast cancer by race and ethnicity: update 2006, CA Cancer J Clin 56 (2006), pp. 168–183. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (127)
13 B. DiNubila, E. Cassano and L. Urban et al., Radiologic features and pathological-biological correlations in 348 women with breast cancer under 35 years old, Breast 2 (1016) (2006).
14 S. Weber-Mangal, H.P. Sinn and S. Popp et al., Breast cancer in young women (<35 years): genomic aberrations detected by comparative genomic hybridization, Int J Cancer 107 (2003), pp. 583–592. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
15 American Cancer Society, Breast cancer facts and figures http://www.cancer.org/downloads/stt/caff2005brf.pdf Accessed April 8, 2008.
16 A. Bleyer, A. Viny and R. Barr, Cancer in 15−29 year olds by primary site, Oncologist 11 (2006), pp. 590–601. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
17 R.E. Tarone, Breast cancer trends among young women in the United States, Epidemiology 17 (2006), pp. 588–590. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
18 Anonymous , Stat bite: Breast cancer in young US women, J Nat Cancer Inst 98 (2006), p. 1762.
19 V. Kaklamani, A genetic signature can predict prognosis and response to therapy in breast cancer: oncotype DX, Expert Rev Mol Diagn 6 (2006), pp. 803–809. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)
20 N. Klauber-DeMore, Tumor biology of breast cancer in young women, Breast Dis 23 (2005−2006), pp. 9–15.
21 M.J. van de Vijver, H.D. He and L.J. van't Veer et al., A gene expression signature as a predictor of survival in breast cancer, N Engl J Med 347 (2002), pp. 1999–2009. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1754)
22 S. Braun, F.D. Vogl and B. Naume et al., A pooled analysis of bone marrow micrometastases in breast cancer, N Engl J Med 353 (2005), pp. 793–802. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (216)
23 K. Amend, D. Hicks and C.B. Ambrosone, Review: Breast cancer in African-American women: differences in tumor biology from European-American women, Cancer Res 66 (2006) 8327–8230.
24 J.A. van der Hage, C.J.H. van ed Velde and H. Putter et al., Impact of established prognostic factors in early-stage breast cancer in very young breast cancer patients: a translational research project using pooled datasets derived from 4 EORTC breast group trials: Abstract 6024, Breast Cancer Research and Treatment 100 (2006), p. S264.
25 M.A. Bollet, B. Sigal-Zafrani and V. Mazeau et al., Age remains the first prognostic factor for loco-regional recurrence in young (<40 years) women treated with breast conserving surgery first, Radiother Oncol 82 (2007), pp. 272–280. Article | PDF (119 K) | View Record in Scopus | Cited By in Scopus (16)
26 G. MacKarem, C.A. Roche and K.S. Hughes, The effectiveness of the Gail Model in estimating risk for development of breast cancer in women under 40 years of age, Breast J 7 (2001), pp. 34–39. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
27 J. Chen, D. Pee and R. Ayyagari et al., Projecting absolute invasive breast cancer risk in white women with a model that includes mammographic density, J Nat; Cancer Inst 98 (2006), pp. 1215–1226. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (58)
28 N. Loman, O. Johannsson and U. Kristoffersson et al., Family history of breast and ovarian cancers and BRCA1and BRCA2 Mutations in a population-based series of early-onset breast cancer, J Nat Cancer Inst 93 (2001), pp. 1215–1223. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (84)
29 L.A. Newman, Breast cancer in African-American women, Oncologist 10 (2005), pp. 1–14. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (30)
30 D.M. Winn, The Long Island Breast Cancer Study Project, Nat Rev Cancer 5 (2005), pp. 986–994. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
31 A.L. Herbst, H. Ulfelder and D.C. Poskanzer, Adenocarcinoma of the vagina: Association of maternal stilbestrol use with tumor appearance in young women, N Engl J Med 284 (1971), pp. 878–881. View Record in Scopus | Cited By in Scopus (541)
32 S. Schrager and B.E. Potter, Diethylstilbestrol exposure, Am Fam Physician 69 (2004), pp. 2395–2440.
33 T. Colton, E.R. Geenberg and L. Noller et al., Breast cancer in mothers prescribed diethylstilbestrol in pregnancy: Further follow-up, JAMA 269 (1993), pp. 2096–3100.
34 J. Prescott, M. Huiyan, L. Bernstein and G. Ursin, Cigarette smoking is not associated with breast cancer risk in young women, Cancer Epidemiol Biomarkers Prev 16 (2007), pp. 620–622. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
35 D.L. Wahner-Roedler and I.A. Petersen, Risk of breast cancer and breast cancer characteristics in women after treatment for Hodgkin's lymphoma, Drugs Today 40 (2004), pp. 865–879. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
36 D. Longo, Radiation in Hodgkin's disease, J Nat Cancer Inst 97 (2005), pp. 1394–1395. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
37 L. Diller, C.M. Nancarrow and K. Shaffer et al., Breast cancer screening in women previously treated for Hodgkin's disease: a prospective cohort study, J Clin Oncol 20 (2002), pp. 2085–2091. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (45)
38 G. Ursin, R.K. Ross and J. Sullivan-Halley et al., Use of oral contraceptives and the risk of breast cancer in young women, Breast Cancer Res Treat 50 (1998), pp. 175–184. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
39 S.A.N. Silvera, A.B. Miller and T.E. Rohan, Oral contraceptive use and risk of breast cancer among women with a family history of breast cancer: a prospective, cohort study, Cancer Causes Control 16 (2005), pp. 1059–1063. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
40 V. Cogliano, Y. Grosse and R. Baan et al., Carcinogenicity of combined oestrogen-progestagen contraceptives and menopausal treatment, Lancet Oncol 6 (2005), pp. 552–553. Article | PDF (45 K) | View Record in Scopus | Cited By in Scopus (60)
41 K.E. Innes and T.E. Byers, First pregnancy characteristics and subsequent breast cancer risk among young women, Int J Cancer 112 (2004), pp. 306–311. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
42 V. Beral, D. Bull and R. Doll et al., Collaborative Group on Hormonal Factors in Breast Cancer, Lancet Oncol 363 (2004), pp. 1007–1016.
43 K.B. Michels, F. Xue, G.A. Colditz and W.C. Willett, Induced and spontaneous abortion and incidence of breast cancer among young women, Arch Intern Med 167 (2007), pp. 814–820. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
44 M. Ahlgren, T. Sorensen and J. Wohlfardt et al., Birth weight and risk of breast cancer in a cohort of 106, 504 women, Int J Cancer 107 (2003), pp. 997–1000. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (52)
45 M.R. Forman, M.M. Cantwell, C. Ronckers and Y. Zhang, Through the looking glass at early-life exposures and breast cancer risk, Cancer Invest 23 (2005), pp. 609–624. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
46 W.D. Thompson and D.T. Janerich, Maternal age at birth and risk of breast cancer in daughters, Epidemiology 1 (1990), pp. 101–106. View Record in Scopus | Cited By in Scopus (30)
47 R.S. Taylor, B.E. Carroll and J.W. Lloyd, Mortality among women in three Catholic orders with special references to cancer, Cancer 12 (1959), pp. 1207–1225. Full Text via CrossRef
48 J.R. Palmer, L.A. Wise and E.E. Hatch et al., Prenatal diethylstilbestrol exposure and risk of breast cancer, Cancer Epidemiol Biomarkers Prev 15 (2006), pp. 1509–1514. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (53)
49 M. Salhab, W. Al Sarakbi and K. Mokbel, In vitro fertilization and breast cancer risk: a review, Int J Fertil Womens Med 50 (2005), pp. 259–266. View Record in Scopus | Cited By in Scopus (13)
50 D. Mintzer, J. Glassburn, B.A. Mason and D. Sataloff, Breast cancer in the very young patient, Oncologist 7 (2002), pp. 547–554. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
51 M. Golshan, A. Miron and A.J. Nixon et al., The prevalence of germline BRCA1 and BRCA2 mutations in young women with breast cancer undergoing breast conservation therapy, Am J Surg 192 (2006), pp. 58–62. Article | PDF (77 K) | View Record in Scopus | Cited By in Scopus (6)
52 K. Heimdal, L. Maehle and J. Apold et al., The Norwegian founder mutations in BRCA1: high penetrance confirmed in an incident cancer series and differences observed in the risk of ovarian cancer, Eur J Cancer 39 (2003), pp. 2205–2213. Article | PDF (218 K) | View Record in Scopus | Cited By in Scopus (14)
53 S.N. Stacey, P. Sulem and O.T. Jonannson et al., The BARD1Cys557Ser Variant and breast cancer risk in Iceland, PLoS Med 3 (2006), p. e217. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
54 M.D. Reeves, T.M. Yawitch and N.C. van der Merve et al., BRCA1 mutations in South African breast and/or ovarian cancer families: evidence of a novel founder mutation in Afrikaner families, Int J Cancer 110 (2004), pp. 677–682. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
55 A. Antoniou, P.D. Pharoah and S.A. Narod et al., Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case series unselected for family history: a combined analysis of 22 studies, Am J Hum Genet 72 (2003), pp. 1117–1130. Article | PDF (524 K) | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (550)
56 Office of National Statistics, Cancer trends in England and Wales: 1950–1999, Studies on Medical and Population Subjects 66 (2001).
57 A. Broeks, L.M. Braaf and A. Huseinovic et al., Identification of women with an increased risk of developing radiation-induced breast cancer: a case only study, Breast Cancer Res 9 (2007), p. R26. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)
58 D.B. Thomas, D.L. Gao and R.M. Ray et al., Randomized trial of breast self-examination in Shanghai: Final results, J Natl Cancer Inst 94 (2002), pp. 1445–1457. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (205)
59 American Society of Clinical Oncology, Statement of the American Society of Clinical Oncology: genetic testing for cancer susceptibility, J Clin Oncol 14 (1996), pp. 1730–1736.
60 V.J. Harris and V.P. Jackson, Indications for breast imaging in women under age 35 years, Radiology 172 (1989), pp. 445–448. View Record in Scopus | Cited By in Scopus (11)
61 N. Houssami, L. Irwig and J.M. Simpson et al., Sydney breast imaging accuracy study: comparative sensitivity and specificity of mammography and sonography in young women with symptoms, Am J Roentgenol 180 (2003), pp. 935–940. View Record in Scopus | Cited By in Scopus (38)
62 M. Murrow, S. Wong and L. Ventra, The evaluation of breast masses in women younger than 40 years of age, Surgery 124 (1998), pp. 634–641.
63 E.D. Pisano, C. Gatsonis and E. Hendrick et al., Diagnostic performance of digital versus film mammography for breast cancer screening, N Engl J Med 353 (2005), pp. 1773–1783. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (312)
64 C.D. Lehman, C. Gatsonis and C.K. Kuhl et al., MRI evaluation of the contralateral breast in women with recently diagnosed breast cancer, N Engl J Med 356 (2007), pp. 1295–1305.
65 M. Kriege, C.T. Brekelmans and I.M. Obdeijn et al., Factors affecting sensitivity and specificity of screening mammography and MRI in women with inherited risk for breast cancer, Breast Cancer Res Treat 100 (2006), pp. 109–119. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
66 S.K. Plevritis, A.W. Kurian and B.M. Sigal, Cost-effectiveness of screening BRCA1/2 mutation carriers with breast magnetic resonance imaging, JAMA 295 (2006), pp. 2374–2384. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
67 J.A. Smith and E. Andreopoulou, An overview of the status of screening technology for breast cancer, Ann Oncol 15 (2004), pp. 18–26.
68 I. Reiser, R.M. Nishikawa and M.L. Giger et al., Computerized mass detection for digital breast tomosynthesis directly from the projection images, Med Phys 33 (2006), pp. 482–491. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (27)
69 M. Brennan, J. French and N. Houssami et al., Breast cancer in young women, Aust Fam Physician 34 (2005), pp. 851–855. View Record in Scopus | Cited By in Scopus (8)
70 H.I. Vargas, M.P. Vargas and K. Eldrageely et al., Outcomes of surgical and sonographic assessment of breast masses in women younger than 30, Am Surg 71 (2005), pp. 716–719. View Record in Scopus | Cited By in Scopus (2)
71 K.A. Skinner, H. Silberman, R. Sposto and M.J. Silverstein, Palpable breast cancers are inherently different from nonpalpable breast cancers, Ann Surg Oncol 8 (2001), pp. 705–710. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)
72 B. Fisher, J.P. Constantino and D.L. Wickerham et al., Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 Study, J Natl Cancer Inst 97 (2005), pp. 1652–1662. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (200)
73 M.C. King, J.H. Marks, J.B. Mandell and N.Y.B.C.S. Group, Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2, Science 302 (2003), pp. 643–646. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (425)
74 S.M. Domchek, J.E. Stopfer and T.R. Rebbeck, Bilateral risk-reducing oophorectomy in BRCA1 and BRCA2 carriers, J Natl Compr Canc Netw 4 (2006), pp. 177–182. View Record in Scopus | Cited By in Scopus (3)
75 T.R. Rebbeck, T. Friebel and T. Wagner et al., Effect of short-term hormone replacement therapy on breast cancer risk reduction after bilateral prophylactic oophorectomy in BRCA1 and BRCA2 mutation carriers: the PROSE Study Group, J Clin Oncol 23 (2005), pp. 7804–7810. View Record in Scopus | Cited By in Scopus (77)
76 L. Lostumbo, N. Carbine, J. Wallace and J. Ezzo, Prophylactic mastectomy for the prevention of breast cancer, Cochrane Database Syst Rev (2004) October 18;(4):CD002748.
77 P. Hopwood, A. Lee and A. Shenton et al., Clinical follow-up after bilateral risk-reducing (propylactic) mastectomy: mental health and body image outcomes, Psychooncology 9 (2000), pp. 462–472. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (40)
78 K.A. Metcalfe, M.J. Esplen, V. Goel and S.A. Narod, Psychosocial functioning in women who have undergone bilateral prophylactic mastectomy, Psychooncology 13 (2004), pp. 14–25. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)
79 M.B. Hatcher, L. Fallowfield and R. A'Hern, The psychosocial impact of bilateral prophylactic mastectomy: Prospective study using questionnaires and semistructured interviews, Br Med J 322 (2001), p. 76. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (61)
80 I.L. Wapnir and S.J. Anderson, Prognosis after ipsilateral breast cancer recurrence and locoregional recurrences in five National Surgical Adjuvant Breast and Bowel Project node-positive adjuvant breast cancer trials, J Clin Oncol 24 (2006), pp. 2028–2037. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (44)
81 P. Zhou, S. Gautam and A. Recht, Factors affecting outcome for young women with early stage breast cancer treated with breast-conserving therapy, Breast Cancer Res Treat 101 (2007), pp. 51–57. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
82 A.C. Voogd, M. Nielsen and J.L. Peterse et al., Differences in risk factors for local and distant recurrence after breast conserving therapy or mastectomy for Stage I and Stage II breast cancer: pooled results of two large European randomized trials, J Clin Oncol 19 (2001), pp. 1688–1697. View Record in Scopus | Cited By in Scopus (158)
83 G.H. de Bock, J.A. van der Hage and H. Putter et al., Isolated loco-regional recurrences of breast cancer is more common in young patients and following breast conserving therapy: long-term results of European Organisation for Research and Treatment of Cancer studies, Eur J Cancer 42 (2006), pp. 351–356. Article | PDF (120 K) | View Record in Scopus | Cited By in Scopus (20)
84 K.J. Van Zee, L. Liberman and B. Samli et al., Long-term follow-up of women with ductal carcinoma in situ treated with breast-conserving surgery, Cancer 86 (1999), pp. 1757–1767. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (68)
85 M. van der Leest, L. Evers and M.J.C. van der Sangen et al., The safety of breast-conserving therapy in patients with breast cancer aged ≤40 years, Cancer 109 (2007), pp. 1957–1964. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
86 C. Seynaeve, L.M. Van Den Bosch and C.T. Brekelmans et al., Local recurrence following lumpectomy and irradiation in familial and hereditary vs. sporadic breast cancer patients [abstract], Proc ASCO 17 (1998), p. 119a.
87 V.G. Vogel, J.P. Costantino and D.L. Wickerham et al., Effects of tamoxifen versus raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene(STAR) P-2 trial, JAMA 295 (2006), pp. 2727–2741. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (340)
88 V.C. Jordan, Chemoprevention of breast cancer with selective oestrogen-receptor modulators, Nat Rev Cancer 7 (2007), pp. 46–53. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (27)
89 R. Jackesz, H. Hausmaninger and E. Kubista et al., Randomized adjuvant trial of tamoxifen and goserelin versus cyclophosphamide, methotrexate, and fluorouracil: evidence for the superiority of treatment with endocrine blockade in premenopausal women with hormone-responsive breast cancer-Austrian Breast and Colorectal Study Group Trial 5, J Clin Oncol 20 (2002), pp. 4621–4627.
90 M. Gnant, R. Greil and E. Kubista et al., The impact of treatment-induced amenorrhea on survival of premenopausal patients with endocrine-responsive breast cancer: 10-year results of ABCSG-05 (CMF vs. goserelin+tamoxifen): Abstract 17, Breast Cancer Res Treat 100 (2006), p. S11.
91 A.H. Boekhout, J.H. Beijnen and J.H.M. Schellens, Symptoms and treatment of cancer-therapy induced early menopause, Oncologist 11 (2006), pp. 641–654. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)
92 M.T. Knobf, The influence of endocrine effects of adjuvant therapy on quality of life outcomes in the young breast cancer survivor, Oncologist 11 (2006), pp. 96–110. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
93 C.A. Cullinane, J. Lubinski and S.L. Neuhausen et al., Effect of pregnancy as a risk factor for breast cancer in BRCA1/BRCA2 mutation carriers, Int J Cancer 117 (2005), pp. 988–991. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (40)
94 H. Jernstrom, C. Lerman and P. Ghadirian et al., Pregnancy and the risk of early breast cancer in carriers of BRCA1 and BRCA2, Lancet Oncol 354 (1999), pp. 1846–1850. Article | PDF (78 K) | View Record in Scopus | Cited By in Scopus (119)
95 D.T. Janerich and M.B. Hoff, Evidence for cross-over in breast cancer risk factors, Am J Epidemiol 116 (1982), pp. 737–742. View Record in Scopus | Cited By in Scopus (46)
96 S. Loibl, G. von Minckwitz and K. Gwyn et al., Breast carcinoma during pregnancy: International recommendations from an expert meeting, Cancer 106 (2006), pp. 237–246. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (44)
97 L.P. Middleton, M. Amin and K. Gwyn et al., Breast carcinoma in pregnant women: assessment of clinicopathologic and immunohistochemical features, Cancer 98 (2005), pp. 1055–1060.
98 J.R. Daling, K.E. Malone and D.R. Doody et al., The relation of reproductive factors to mortality from breast cancer, Cancer Epidemiol Biomarkers Prev 11 (2002), pp. 235–241. View Record in Scopus | Cited By in Scopus (32)
99 P. Schedin, Pregnancy-associated breast cancer and metastases, Nat Rev Cancer 6 (2006), pp. 281–291. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (33)
100 H. Kuerer, K. Gwyn, F.C. Ames and R. Theriault, Conservative surgery and chemotherapy for breast carcinoma during pregnancy, Surgery 131 (2002), pp. 108–110. Article | PDF (39 K) | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
101 F. Amersi and N.M. Hansen, The benefits and limitations of sentinel node biopsy, Curr Treat Options Oncol 7 (2006), pp. 141–151. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (14)
102 M.M. Mondi, R.E. Cuenca and D.W. Ollila et al., Sentinel lymph node biopsy during pregnancy: initial clinical experience, Ann Surg Oncol 14 (2007), pp. 218–221. View Record in Scopus | Cited By in Scopus (10)
103 D.L. Berry, R.L. Theriault and F.A. Holmes et al., Management of breast cancer during pregnancy using a standardized protocol, J Clin Oncol 17 (1999), pp. 855–861. View Record in Scopus | Cited By in Scopus (146)
104 M.E.K. Hahn, P.H. Johnson and N. Gordon et al., Treatment of pregnant breast cancer patients and outcomes of children exposed to chemotherapy in utero, Cancer 107 (2006), pp. 1219–1226.
105 B.F. Hankey, B. Miller, R. Curtis and C. Kosary, Trends in breast cancer in younger women in contrast o older women, J Nat; Cancer Inst Monogr 16 (1994), pp. 7–14. View Record in Scopus | Cited By in Scopus (77)
106 E. Von Shultz, H. Johansson, N. Wilking and L.E. Rutqvist, Influence of prior and subsequent pregnancy on breast cancer prognosis, J Clin Oncol 13 (1995), pp. 430–434.
107 K. Calhoun and N. Hansen, The effect of pregnancy on survival in women with a history of breast cancer, Breast Dis 23 (2005−2006), pp. 81–86.
108 S. Geller, A.S. Coates and A. Goldhirsch et al., Effect of pregnancy on overall survival after the diagnosis of early-stage breast cancer, J Clin Oncol 19 (2001), pp. 1671–1675.
109 S. Shousha, Breast carcinoma presenting during or shortly after pregnancy and lactation, Arch Pathol Lab Med 124 (2000), pp. 1053–1060. View Record in Scopus | Cited By in Scopus (26)
110 M. Mahue-Giangreco, G. Ursin, J. Sullivan-Halley and L. Bernstein, Induced abortion, miscarriage, and breast cancer risk of young women, Cancer Epidemiol Biomarkers Prev 12 (2003), pp. 209–214. View Record in Scopus | Cited By in Scopus (14)
111 M. Sonmezer and K. Oktay, Fertility preservation in young women undergoing breast cancer therapy, Oncologist 11 (2006), pp. 422–434. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (40)
112 D.H. Baucom, L.S. Porter and J.S. Kirby et al., Psychosocial issues confronting young women with breast cancer, Breast Dis 23 (2005−2006), pp. 103–113.
113 P. Fobair, S.L. Stewart and S. Chang et al., Body image and sexual problems in young women with breast cancer, Psychooncology 15 (2006), pp. 579–594. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
114 T.M. Kolb, J. Lichy and J.H. Newhouse, Comparison of the performance of screening mammography, physical exam, and breast ultrasound, and analysis of features that influence them: an analysis of 27,825 patient evaluations, Radiology 225 (2002), pp. 165–175. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (296)
  相似文献   

2.
Recovery after a craniotomy procedure is an important step for the success of a neurosurgical procedure. During this process, it is necessary to pursue all of the objectives of current neurosurgical anaesthesia: maintain brain homeostasis, avoid oedema formation (hyponatremia, seizures, steroids withdrawal), control intracranial pressure (normocapnia, normothermia, prevent shivering and nausea or vomiting), prevent and treat pain.Immediate post-procedural neurological evaluation is mandatory to detect any postoperative cerebral complication as soon as possible. If the patient does not obey command, extubation has to be delayed. After posterior fossa procedures, intact cranial nerve functions must be confirmed before extubation.If extubation is not possible and a delayed recovery is indicated, intracranial pressure monitoring may become useful. “Sedation windows” are needed in order to perform neurological evaluations at regular, predefined time points for detection of complications and for minimizing the duration of mechanical ventilation.A brain CT should be performed rapidly when a unexpected neurological deficit or change in consciousness have evolved. In many neurosurgical centers, a systematic CT-scan is performed in the first 24-hours after surgery in order to detect a clinically silent neurosurgical complication.Even if an intensive care unit is not available after a craniotomy procedure, central neurosurgical patients require an intensive neurological and haemodynamic monitoring at least for the time period of one night after surgery. Neurosurgery and anaesthesia experts should be immediately available over 24-hours every day in case of severe complications.
• Provide "stress-free recovery” for central neurosurgical patients
• Avoid hypertension, hypercapnia, shivering, pain and coughing
• Perform neurological examination as soon as possible after surgery
• A neurological score is recommended in order to improve follow-up.
• Maintain normothermia, normotension, euvolemia and normoosmolality
• Closely and continuously monitor over at least 24 hours after surgery
• Perform CT or MR immediately if neurological deterioration or seizures occur
• Control the ICP if postoperative sedation becomes necessary
• Work on anaesthetic techniques and monitoring that allows for intra- and post-operative homeostasis is needed, and research to minimize anaesthesia- and procedure-related sequelae is warranted
• Finding a good neurological score to be applied on postoperative craniotomy patients and for improving the detection of cerebral complications would be helpful
• Identifying markers of postcraniotomy brain damage to guide therapy would be useful

References

1 K.A. Miller, C.P. Harkin and P.L. Bailey, Postoperative tracheal extubation, Anesthesia and Analgesia 80 (1995), pp. 149–172. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (64)
2 S.M. Frank, L.A. Fleisher and K.F. Olson et al., Multivariate determinants of early postoperative oxygen consumption in elderly patients. Effects of shivering, body temperature, and gender, Anesthesiology 83 (1995), pp. 241–249. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (83)
3 S.M. Frank, M.S. Higgins and M.J. Breslow et al., The catecholamine, cortisol, and hemodynamic responses to mild perioperative hypothermia. A randomized clinical trial, Anesthesiology 82 (1995), pp. 83–93. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (179)
4 S. Motamed, K. Klubien and M. Edwardes et al., Metabolic changes during recovery in normothermic versus hypothermic patients undergoing surgery and receiving general anesthesia and epidural local anesthetic agents, Anesthesiology 88 (1998), pp. 1211–1218. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
5 R.L. Lennon, M.P. Hosking and M.A. Conover et al., Evaluation of a forced-air system for warming hypothermic postoperative patients, Anesthesia and Analgesia 70 (1990), pp. 424–427. View Record in Scopus | Cited By in Scopus (34)
6 A. Sharkey, J.M. Lipton and M.T. Murphy et al., Inhibition of postanesthetic shivering with radiant heat, Anesthesiology 66 (1987), pp. 249–252. View Record in Scopus | Cited By in Scopus (34)
7 M.J. Breslow, S.D. Parker and S.M. Frank et al., Determinants of catecholamine and cortisol responses to lower extremity revascularization. The PIRAT Study Group, Anesthesiology 79 (1993), pp. 1202–1209. View Record in Scopus | Cited By in Scopus (71)
8 C.M. Moore, M.H. Cross and J.P. Desborough et al., Hormonal effects of thoracic extradural analgesia for cardiac surgery, British Journal of Anaesthesia 75 (1995), pp. 387–393. View Record in Scopus | Cited By in Scopus (79)
9 M. Yokoyama, Y. Itano and H. Katayama et al., The effects of continuous epidural anesthesia and analgesia on stress response and immune function in patients undergoing radical esophagectomy, Anesthesia and Analgesia 101 (2005), pp. 1521–1527. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
10 A. Gottschalk, L.C. Berkow and R.D. Stevens et al., Prospective evaluation of pain and analgesic use following major elective intracranial surgery, Journal of Neurosurgery 106 (2007), pp. 210–216. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
*11 F. Bilotta, R. Caramia and F.P. Paoloni et al., Early postoperative cognitive recovery after remifentanil-propofol or sufentanil-propofol anaesthesia for supratentorial craniotomy: a randomized trial, European Journal of Anaesthesiology 24 (2007), pp. 122–127. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
*12 K. Gerlach, T. Uhlig and M. Huppe et al., Remifentanil-propofol versus sufentanil-propofol anaesthesia for supratentorial craniotomy: a randomized trial, European Journal of Anaesthesiology 20 (2003), pp. 813–820. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
13 J. Guy, B.J. Hindman and K.Z. Baker et al., Comparison of remifentanil and fentanyl in patients undergoing craniotomy for supratentorial space-occupying lesions, Anesthesiology 86 (1997), pp. 514–524. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (109)
14 S.H. Lim, N.M. Chin and H.Y. Tai et al., Prophylactic esmolol infusion for the control of cardiovascular responses to extubation after intracranial surgery, Annals of the Academy of Medicine, Singapore 29 (2000), pp. 447–451. View Record in Scopus | Cited By in Scopus (7)
15 M.M. Todd, D.S. Warner and M.D. Sokoll et al., A prospective, comparative trial of three anesthetics for elective supratentorial craniotomy. Propofol/fentanyl, isoflurane/nitrous oxide, and fentanyl/nitrous oxide, Anesthesiology 78 (1993), pp. 1005–1020. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (108)
16 Y. Inagaki, H. Shindo and T. Mashimo et al., The effects of epidural fentanyl on hemodynamic responses during emergence from isoflurane anesthesia and tracheal extubation: a comparison with intravenous fentanyl, Anesthesia and Analgesia 85 (1997), pp. 328–335. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
17 K. Nishina, K. Mikawa and N. Maekawa et al., Fentanyl attenuates cardiovascular responses to tracheal extubation, Acta Anaesthesiologica Scandinavica 39 (1995), pp. 85–89. View Record in Scopus | Cited By in Scopus (24)
18 J. Conti and D. Smith, Haemodynamic responses to extubation after cardiac surgery with and without continued sedation, British Journal of Anaesthesia 80 (1998), pp. 834–836. View Record in Scopus | Cited By in Scopus (27)
19 A. Kulkarni, G. Price and M. Saxena et al., Difficult extubation: calming the sympathetic storm, Anaesthesia and Intensive Care 32 (2004), pp. 413–416. View Record in Scopus | Cited By in Scopus (7)
20 A. Basali, E. Mascha and I. Kalfas et al., Relation between perioperative hypertension and intracranial hemorrhage after craniotomy, Anesthesiology 93 (2000), pp. 48–54. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (39)
21 R.M.J. Bryan, Cerebral blood flow and energy metabolism during stress, The American Journal of Physiology 259 (1990), pp. H269–H280. View Record in Scopus | Cited By in Scopus (42)
22 N. Bruder, D. Pellissier and P. Grillo et al., Cerebral hyperemia during recovery from general anesthesia in neurosurgical patients, Anesthesia and Analgesia 94 (2002), pp. 650–654. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)
23 R.A. Falero, A.L. Pereda and I.F. Rodriguez, Transient hyperemia immediately after rapid decompression of chronic subdural hematoma, Neurosurgery 47 (2000), pp. 1468–1469. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
24 C.H. Kim, Transient hyperemia immediately after rapid decompression of chronic subdural hematoma, Neurosurgery 47 (2000), pp. 791–792. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
25 P.M. Meyers, C.C. Phatouros and R.T. Higashida, Hyperperfusion syndrome after intracranial angioplasty and stent placement, Stroke 37 (2006), pp. 2210–2211. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
26 K. Ogasawara, K. Koshu and T. Yoshimoto et al., Transient hyperemia immediately after rapid decompression of chronic subdural hematoma, Neurosurgery 45 (1999), pp. 484–488 Discussion 488–489. Full Text via CrossRef
27 T. Pfefferkorn, T. Mayer and S. Von Stuckrad-Barre et al., Hyperperfusion-induced intracerebral hemorrhage after carotid stenting documented by TCD, Neurology 57 (2001), pp. 1933–1935. View Record in Scopus | Cited By in Scopus (15)
28 N.J. Bruder, Awakening management after neurosurgery for intracranial tumours, Current Opinion in Anaesthesiology 15 (2002), pp. 477–482. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
29 J. Brucia and E. Rudy, The effect of suction catheter insertion and tracheal stimulation in adults with severe brain injury, Heart and Lung 25 (1996), pp. 295–303. Abstract | PDF (1958 K) | View Record in Scopus | Cited By in Scopus (18)
30 M. Gemma, C. Tommasino and M. Cerri et al., Intracranial effects of endotracheal suctioning in the acute phase of head injury, Journal of Neurosurgical Anesthesiology 14 (2002), pp. 50–54. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
31 M. Leone, J. Albanese and X. Viviand et al., The effects of remifentanil on endotracheal suctioning-induced increases in intracranial pressure in head-injured patients, Anesthesia and Analgesia 99 (2004), pp. 1193–1198 table of contents. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
32 S. Constantini, S. Cotev and Z. Rappaport et al., Intracranial pressure monitoring after elective intracranial surgery. A retrospective study of 514 consecutive patients, Journal of Neurosurgery 69 (1988), pp. 540–544. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)
33 J. Vassilouthis, S. Anagnostaras and A. Papandreou et al., Is postoperative haematoma an avoidable complication of intracranial surgery, British Journal of Neurosurgery 13 (1999), pp. 154–157. View Record in Scopus | Cited By in Scopus (6)
34 R.M. Lazar, B.F. Fitzsimmons and R.S. Marshall et al., Midazolam challenge reinduces neurological deficits after transient ischemic attack, Stroke 34 (2003), pp. 794–796. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
35 R.M. Lazar, B.F. Fitzsimmons and R.S. Marshall et al., Reemergence of stroke deficits with midazolam challenge, Stroke 33 (2002), pp. 283–285. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (32)
36 G.D. Thal, M.D. Szabo and M. Lopez-Bresnahan et al., Exacerbation or unmasking of focal neurologic deficits by sedatives, Anesthesiology 85 (1996), pp. 21–25 Discussion 29A-30A. Full Text via CrossRef
37 N. Bruder, J. Stordeur and P. Ravussin et al., Metabolic and hemodynamic changes during recovery and tracheal extubation in neurosurgical patients: immediate versus delayed recovery, Anesthesia and Analgesia 89 (1999), pp. 674–678. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
38 G. Carrasco, R. Molina and J. Costa, Propofol vs midazolam in short-, medium-, and long-term sedation of critically ill patients: A cost benefit analysis, Chest 103 (1993), pp. 557–564. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (167)
39 C. Chamorro, F.J. de Latorre and A. Montero et al., Comparative study of propofol versus midazolam in the sedation of critically ill patients: results of a prospective, randomized, multicenter trial see comments, Critical Care Medicine 24 (1996), pp. 932–939. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (96)
40 J.P. Kress, M.F. O'Connor and A.S. Pohlman et al., Sedation of critically ill patients during mechanical ventilation. A comparison of propofol and midazolam, American Journal of Respiratory and Critical Care Medicine 153 (1996), pp. 1012–1018. View Record in Scopus | Cited By in Scopus (91)
41 F. Snellen, P. Lauwers and R. Demeyere et al., The use of midazolam versus propofol for short-term sedation following coronary artery bypass grafting, Intensive Care Medicine 16 (1990), pp. 312–316. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (45)
42 B. Muellejans, T. Matthey and J. Scholpp et al., Sedation in the intensive care unit with remifentanil/propofol versus midazolam/fentanyl: a randomised, open-label, pharmacoeconomic trial, Critical Care 10 (2006), p. R91. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (0)
43 A. Schubert, E.J. Mascha and E.L. Bloomfield et al., Effect of cranial surgery and brain tumor size on emergence from anesthesia, Anesthesiology 85 (1996), pp. 513–521. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
*44 W.C. Ziai, P.N. Varelas and S.L. Zeger et al., Neurologic intensive care resource use after brain tumor surgery: an analysis of indications and alternative strategies, Critical Care Medicine 31 (2003), pp. 2782–2787. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
*45 W. Heinke, S. Zysset and M. Hund-Georgiadis et al., The effect of esmolol on cerebral blood flow, cerebral vasoreactivity, and cognitive performance: a functional magnetic resonance imaging study, Anesthesiology 102 (2005), pp. 41–50. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
46 P. Grillo, N. Bruder and P. Auquier et al., Esmolol blunts the cerebral blood flow velocity increase during emergence from anesthesia in neurosurgical patients, Anesthesia and Analgesia 96 (2003), pp. 1145–1149. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)
47 D. Muzzi, S. Black and T. Losasso et al., Labetalol and esmolol in the control of hypertension after intracranial surgery, Anesthesia and Analgesia 70 (1990), pp. 68–71. View Record in Scopus | Cited By in Scopus (42)
48 C.S. Tietjen, P.D. Hurn and J.A. Ulatowski et al., Treatment modalities for hypertensive patients with intracranial pathology: options and risks, Critical Care Medicine 24 (1996), pp. 311–322. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (51)
49 C. Hormann, G. Luz and J. Langmayr et al., Influence of urapidil on cerebrospinal fluid pressure in humans with uncompromised intracranial compliance, Intensive Care Medicine 21 (1995), pp. 853–855. View Record in Scopus | Cited By in Scopus (4)
50 G. Singbartl and G. Metzger, Urapidil-induced increase of the intracranial pressure in head-trauma patients, Intensive Care Medicine 16 (1990), pp. 272–274. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
51 J.I. Suarez, O.O. Zaidat and M.F. Suri et al., Length of stay and mortality in neurocritically ill patients: impact of a specialized neurocritical care team, Critical Care Medicine 32 (2004), pp. 2311–2317. View Record in Scopus | Cited By in Scopus (45)
52 G. Teasdale and B. Jennet, Assessment of coma and impaired consciousness. A practical scale, Lancet 2 (1974), pp. 81–84. Abstract | Article | PDF (561 K) | View Record in Scopus | Cited By in Scopus (3078)
*53 J. Spears, K.G. TerBrugge and M. Moosavian et al., A discriminative prediction model of neurological outcome for patients undergoing surgery of brain arteriovenous malformations, Stroke 37 (2006), pp. 1457–1464. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
*54 B.E. Pollock and R.D. Brown, Use of the modified Rankin Scale to assess outcome after arteriovenous malformation radiosurgery, Neurology 67 (2006), pp. 1630–1634. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
55 R.F. Spetzler and N.A. Martin, A proposed grading system for arteriovenous malformation, Journal of Neurosurgery 65 (1986), pp. 476–483. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (532)
56 J.C. van Swieten, P.J. Koudstaal and M.C. Visser et al., Interobserver agreement for the assessment of handicap in stroke patients, Stroke 19 (1988), pp. 604–607. View Record in Scopus | Cited By in Scopus (1045)
57 B. Jennet and M. Bond, Assessment of outcome after severe brain damage, Lancet 1 (1975), pp. 480–484.
58 D.A. Karnofsky, W.H. Abelman, L.F. Craver and J.H. Burchenal, The use of nitrogen mustards in the palliative treatment of carcinoma, Cancer 1 (1948), pp. 634–656. Full Text via CrossRef
59 M. Zetterling and E. Ronne-Engström, High intraoperative blood loss may be a risk factor for postoperative hematoma, Journal of Neurosurgical Anesthesiology 16 (2004), pp. 151–155. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
60 J.E. Starmark, D. Stålhammar and E. Holmgren, The Reaction Level Scale (RLS85). Manual and guidelines, Acta Neurochirurgica 91 (1988), pp. 12–20. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (82)
61 M.F. Folstein, S.E. Folstein and P.R. McHugh, “Mini-mental state”: a practical method for grading the cognitive state of patients for the clinician, Journal of Psychiatric Research 12 (1975), pp. 189–198. Abstract | PDF (635 K) | View Record in Scopus | Cited By in Scopus (23017)
*62 S. Einav, Y. Shoshan and H. Ovadia et al., Early postoperative serum S100β levels predict ongoing brain damage after meningioma surgery: a prospective observational study, Critical Care 10 (6) (2006), pp. 1–10.
63 T. Brott, H.P. Adams and C.P. Olinger et al., Measurements of acute cerebral infarction: a clinical examination scale, Stroke 20 (1989), pp. 864–870. View Record in Scopus | Cited By in Scopus (964)
64 F.B. Young and C.J. Weir, Lees KR, for the GAIN international trial steering committee and investigators. Comparison of the National Institutes of Health Stroke Scale with disability outcome measures in acute stroke trials, Stroke 36 (2005), pp. 2187–2192. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (14)
65 C. Collin, D.T. Wade and S. Davis, The Barthel ADL Index; a reliability study, International Disability Studies 10 (1998), pp. 61–63.
66 E.C. Nemergut, A.S. Dumont, U.T. Barry and E.R. Laws, Perioperative management for patients undergoing transsphenoidal pituitary surgery, Anesthesia and Analgesia 101 (2005), pp. 1170–1181. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
67 H. Eloqayli, J. Cappelen and A. Vik, Acute spontaneous subdural haematoma after transsphenoidal surgery, Acta Neurochirurgica 148 (2006), pp. 587–590. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
68 T.C. Origitano, G.J. Petruzzelli, J.P. Leonetti and D. Vandevender, Combined anterior and anterolateral approaches to the cranial base: complication analysis, avoidance, and management, Neurosurgery 58 (Suppl2) (2006), pp. ONS327–ONS337.
69 H. Prabhakar, P.K. Bithal and A. Grag, Tension pneumocephalus after craniotomy in supine position, Journal of Neurosurgical Anesthesiology 15 (2003), pp. 278–281. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
70 R.J. Komotar, J. Mocco and E.R. Ransom et al., Herniation secondary to critical postcraniotomy cerebrospinal fluid hypovolemia, Neurosurgery 57 (2005), pp. 286–292. Full Text via CrossRef
*71 R.J. Komotar, E.R. Ransom and J. Mocco et al., Critical postcraniotomy cerebrospinal fluid hypovolemia: risk factors and outcome analysis, Neurosurgery 59 (2006), pp. 284–290. Full Text via CrossRef
72 S.M. Russell and P.J. Kelly, Incidence and clinical evolution of postoperative deficits after volumetric stereotactic resection of glial neoplasms involving the supplementary motor area, Neurosurgery 52 (2003), pp. 506–516. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
73 A.M. Mauro, C. Bomprezzi and S. Morresi et al., Prevention of early postoperative seizures in patients with primary brain tumours: preliminary experience with oxcarbazepine, Journal of Neuro-Oncology 81 (2007), pp. 279–285. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
74 N.R. Temkin, Antiepileptogenesis and seizure prevention trials with antiepileptic drugs: meta-analysis of controlled trials, Epilepsia 42 (2001), pp. 515–524. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (137)
75 R. Grossman, S. Sadetzki, R. Spiegelmann and Z. Ram, Haemorrhagic complications and the incidence of asymptomatic bleeding associated with stereotactic brain biopsies, Acta Neurochirurgica 147 (2005), pp. 627–631. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
76 U. Samadani, S. Stein and G. Moonis et al., Stereotactic biopsy of brain stem masses: decision analysis and literature review, Surgical Neurology 66 (2006), pp. 484–491.
*77 M.J. McGirt, G.F. Woodworth and A.L. Coon et al., Independent predictors of morbidity after image-guided stereotactic brain biopsy: a risk assessment of 270 cases, Journal of Neurosurgery 102 (2005), pp. 897–901. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
78 D.K. Binder, G.M. Rau and P.A. Starr, Risk factors for hemorrhage during microelectrode-guided deep brain stimulator implantation for movement disorders, Neurosurgery 56 (2005), pp. 722–732. Full Text via CrossRef
*79 R.B. Khan, P.H. Gutin and S.N. Rai et al., Use of diffusion weighted magnetic resonance imaging in predicting early postoperative outcome of new neurological deficits after brain tumor resection, Neurosurgery 59 (2006), pp. 60–66. Full Text via CrossRef
80 S. Ulmer, T.A. Braga and F.G. Barker et al., Clinical and radiographic features of peritumoral infarction following resection of glioblastoma, Neurology 67 (2006), pp. 1668–1670. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
81 G. Stranjalis, S. Korfias and C. Psachoulia et al., Serum S-100B as an indicator of early postoperative deterioration after meningioma surgery, Clinical Chemistry 51 (2005), pp. 202–207. View Record in Scopus | Cited By in Scopus (9)
  相似文献   

3.
The field of neurohormonal physiology is highly complex. Since vasopressin was isolated in the early 1950s, much has been learnt about its function in health and disease, but we are far from a complete understanding of its different and diverse functions, and have much to discover in terms of the complex interactions between vasopressin and other neurohormones and mediators. The development of specific agonists and antagonists for the different vasopressin receptors in recent years has enabled research to examine their individual and combined roles in physiological homeostasis more precisely, and to propose therapeutic uses for these agents in various common disease processes. Vasopressin and its analogues have also been used in the treatment of diabetes insipidus, bleeding oesophageal varices, shock, particularly septic shock, and cardiopulmonary resuscitation. Further research into the physiological roles of vasopressin, the complex signalling pathways associated with receptor stimulation, and the multiple feedback loops which regulate vasopressin secretion and release will offer improved insight into this fascinating peptide, and provide future targets for therapeutic investigation.
• many aspects of vasopressin's functions remain poorly understood
• further research is needed to improve understanding of the molecular basis of receptor function using newly developed specific vasopressin receptor antagonists
• ongoing studies will help to elucidate the complex mechanisms regulating vasopressin synthesis and release, and the interactions of vasopressin with other neurohormonal and vasoregulatory pathways
• clinical studies are warranted to evaluate further the role of vasopressin receptor antagonists in various disease processes, including congestive heart failure and hyponatraemia

References

1 G. Oliver and E.A. Schaefer, On the physiological action of extract of pituitary body and certain other glandular organs, The Journal of Physiology 18 (1895), pp. 277–279.
2 G.M. Mutlu and P. Factor, Role of vasopressin in the management of septic shock, Intensive Care Medicine 30 (2004), pp. 1276–1291. View Record in Scopus | Cited By in Scopus (75)
3 S. Oshikawa, A. Tanoue and T.A. Koshimizu et al., Vasopressin stimulates insulin release from islet cells through V1b receptors: a combined pharmacological/knockout approach, Molecular Pharmacology 65 (2004), pp. 623–629. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (29)
4 A. Tanoue, S. Ito and K. Honda et al., The vasopressin V1b receptor critically regulates hypothalamic-pituitary-adrenal axis activity under both stress and resting conditions, The Journal of Clinical Investigation 113 (2004), pp. 302–309. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (66)
5 H. Weingartner, P. Gold and J.C. Ballenger et al., Effects of vasopressin on human memory functions, Science 211 (1981), pp. 601–603. View Record in Scopus | Cited By in Scopus (39)
6 I.F. Bielsky, S.B. Hu and X. Ren et al., The V1a vasopressin receptor is necessary and sufficient for normal social recognition: a gene replacement study, Neuron 47 (2005), pp. 503–513. Article | PDF (311 K) | View Record in Scopus | Cited By in Scopus (61)
7 R. Acher, J. Chauvet and M.T. Chauvet, Man and the chimaera. Selective versus neutral oxytocin evolution, Advances in Experimental Medicine and Biology 395 (1995), pp. 615–627.
*8 F.M. de Bree and J.P. Burbach, Structure–function relationships of the vasopressin prohormone domains, Cellular and Molecular Neurobiology 18 (1998), pp. 173–191. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (35)
9 A.H. Sklar and R.W. Schrier, Central nervous system mediators of vasopressin release, Physiological Reviews 63 (1983), pp. 1243–1280. View Record in Scopus | Cited By in Scopus (54)
10 P.H. Baylis, Osmoregulation and control of vasopressin secretion in healthy humans, American Journal of Physiology 253 (1987), pp. R671–R678.
11 W. Feldberg, P.G. Guertzenstein and M. Rocha e Silva Jr., Vasopressin release by nicotine: the site of action, British Journal of Pharmacology 54 (1975), pp. 463–474.
12 D.T. den Ouden and A.E. Meinders, Vasopressin: physiology and clinical use in patients with vasodilatory shock: a review, Netherlands Journal of Medicine 63 (2005), pp. 4–13. View Record in Scopus | Cited By in Scopus (15)
13 P.C. Kam, S. Williams and F.F. Yoong, Vasopressin and terlipressin: pharmacology and its clinical relevance, Anaesthesia 59 (2004), pp. 993–1001. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (33)
*14 L. Bankir, Antidiuretic action of vasopressin: quantitative aspects and interaction between V1a and V2 receptor-mediated effects, Cardiovascular Research 51 (2001), pp. 372–390. View Record in Scopus | Cited By in Scopus (70)
15 E.W. Quillen Jr. and A.W. Cowley Jr., Influence of volume changes on osmolality-vasopressin relationships in conscious dogs, American Journal of Physiology 244 (1983), pp. H73–H79. View Record in Scopus | Cited By in Scopus (4)
16 R.W. Schrier, T. Berl and R.J. Anderson, Osmotic and nonosmotic control of vasopressin release, American Journal of Physiology 236 (1979), pp. F321–F332. View Record in Scopus | Cited By in Scopus (19)
17 G. Leng, C.H. Brown and J.A. Russell, Physiological pathways regulating the activity of magnocellular neurosecretory cells, Progress in Neurobiology 57 (1999), pp. 625–655. Article | PDF (644 K) | View Record in Scopus | Cited By in Scopus (139)
18 V. Bahr, N. Franzen and W. Oelkers et al., Effect of exogenous glucocorticoid on osmotically stimulated antidiuretic hormone secretion and on water reabsorption in man, European Journal of Endocrinology 155 (2006), pp. 845–848. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
19 M. Birnbaumer, Vasopressin receptors, Trends in Endocrinology and Metabolism 11 (2000), pp. 406–410. Article | PDF (185 K) | View Record in Scopus | Cited By in Scopus (114)
20 R.H. Oakley, S.A. Laporte and J.A. Holt et al., Association of beta-arrestin with G protein-coupled receptors during clathrin-mediated endocytosis dictates the profile of receptor resensitization, The Journal of Biological Chemistry 274 (1999), pp. 32248–32257. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (240)
21 C. Barberis, B. Mouillac and T. Durroux, Structural bases of vasopressin/oxytocin receptor function, The Journal of Endocrinology 156 (1998), pp. 223–229. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (110)
22 M. Birnbaumer, A. Seibold and S. Gilbert et al., Molecular cloning of the receptor for human antidiuretic hormone, Nature 357 (1992), pp. 333–335. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (257)
23 B. Chini and M. Manning, Agonist selectivity in the oxytocin/vasopressin receptor family: new insights and challenges, Biochemical Society Transactions 35 (2007), pp. 737–741. View Record in Scopus | Cited By in Scopus (8)
24 S.R. Neves, P.T. Ram and R. Iyengar, G protein pathways, Science 296 (2002), pp. 1636–1639. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (285)
*25 C.L. Holmes, D.W. Landry and J.T. Granton, Science review: vasopressin and the cardiovascular system part 1 – receptor physiology, Critical Care (London, England) 7 (2003), pp. 427–434. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (62)
26 C.P. George, F.H. Messerli and J. Genest et al., Diurnal variation of plasma vasopressin in man, The Journal of Clinical Endocrinology and Metabolism 41 (1975), pp. 332–338. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (62)
27 M. Nadal, Secretory rhythm of vasopressin in healthy subjects with inversed sleep–wake cycle: evidence for the existence of an intrinsic regulation, European Journal of Endocrinology 134 (1996), pp. 174–176. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
28 P. Forrest, Vasopressin and shock, Anaesthesia and Intensive Care 29 (2001), pp. 463–472. View Record in Scopus | Cited By in Scopus (29)
29 T.D. Williams, D. Da Costa and C.J. Mathias et al., Pressor effect of arginine vasopressin in progressive autonomic failure, Clinical Science (London) 71 (1986), pp. 173–178. View Record in Scopus | Cited By in Scopus (10)
30 H.P. Peters, J.H. Robben, P.M. Deen and J.F. Wetzels, Water in health and disease: new aspects of disturbances in water metabolism, Netherlands Journal of Medicine 65 (2007), pp. 325–332. View Record in Scopus | Cited By in Scopus (2)
31 G.L. Robertson, Abnormalities of thirst regulation, Kidney International 25 (1984), pp. 460–469. View Record in Scopus | Cited By in Scopus (26)
32 F.C. Bartter and W.B. Schwartz, The syndrome of inappropriate secretion of antidiuretic hormone, American Journal of Medicine 42 (1967), pp. 790–806. Abstract | PDF (1951 K) | View Record in Scopus | Cited By in Scopus (275)
33 A.S. Multz, Vasopressin dysregulation and hyponatremia in hospitalized patients, Journal of Intensive Care Medicine 22 (2007), pp. 216–223. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
34 A. Rai, A. Whaley-Connell, S. McFarlane and J.R. Sowers, Hyponatremia, arginine vasopressin dysregulation, and vasopressin receptor antagonism, American Journal of Nephrology 26 (2006), pp. 579–589. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)
*35 A. Soupart, P. Gross and J.J. Legros et al., Successful long-term treatment of hyponatremia in syndrome of inappropriate antidiuretic hormone secretion with satavaptan (SR121463B), an orally active nonpeptide vasopressin V2-receptor antagonist, Clinical Journal of the American Society of Nephrology 1 (2006), pp. 1154–1160. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (30)
36 S.R. Goldsmith, The role of vasopressin in congestive heart failure, Cleveland Clinic Journal of Medicine 73 (Suppl 3) (2006), pp. S19–S23.
*37 P.R. Kalra, S.D. Anker and A.J. Coats, Water and sodium regulation in chronic heart failure: the role of natriuretic peptides and vasopressin, Cardiovascular Research 51 (2001), pp. 495–509. View Record in Scopus | Cited By in Scopus (32)
38 M.H. Chin and L. Goldman, Correlates of major complications or death in patients admitted to the hospital with congestive heart failure, Archives of Internal Medicine 156 (1996), pp. 1814–1820. View Record in Scopus | Cited By in Scopus (82)
39 L. Klein, C.M. O'Connor and J.D. Leimberger et al., Lower serum sodium is associated with increased short-term mortality in hospitalized patients with worsening heart failure: results from the Outcomes of a Prospective Trial of Intravenous Milrinone for Exacerbations of Chronic Heart Failure (OPTIME-CHF) study, Circulation 111 (2005), pp. 2454–2460. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (69)
40 S. Kumar, S. Rubin, P.J. Mather and D.J. Whellan, Hyponatremia and vasopressin antagonism in congestive heart failure, Clinical Cardiology 30 (2007), pp. 546–551. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
*41 M.A. Konstam, M. Gheorghiade and J.C. Burnett Jr. et al., Effects of oral tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial, The Journal of the American Medical Association 297 (2007), pp. 1319–1331. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (133)
42 P. Gines, T. Berl and M. Bernardi et al., Hyponatremia in cirrhosis: from pathogenesis to treatment, Hepatology 28 (1998), pp. 851–864. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (89)
43 J.P. Morello, A. Salahpour and A. Laperriere et al., Pharmacological chaperones rescue cell-surface expression and function of misfolded V2 vasopressin receptor mutants, The Journal of Clinical Investigation 105 (2000), pp. 887–895. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (221)
*44 C.L. Holmes, D.W. Landry and J.T. Granton, Science review: vasopressin and the cardiovascular system part 2 – clinical physiology, Critical Care (London, England) 8 (2004), pp. 15–23. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (41)
*45 D.W. Landry, H.R. Levin and E.M. Gallant et al., Vasopressin deficiency contributes to the vasodilation of septic shock, Circulation 95 (1997), pp. 1122–1125. View Record in Scopus | Cited By in Scopus (383)
46 T. Sharshar, A. Blanchard and M. Paillard et al., Circulating vasopressin levels in septic shock, Critical Care Medicine 31 (2003), pp. 1752–1758. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (133)
*47 T. Sharshar, R. Carlier and A. Blanchard et al., Depletion of neurohypophyseal content of vasopressin in septic shock, Critical Care Medicine 30 (2002), pp. 497–500. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (85)
*48 J.L. Vincent, Vasopressin in hypotensive and shock states, Critical Care Clinics 22 (2006), pp. 187–197. Abstract | View Record in Scopus | Cited By in Scopus (6)
  相似文献   

4.
Mild therapeutic hypothermia is an important neuroprotective concept after out-of-hospital cardiac arrest[*37] and [*97] and improves outcome after pediatric ischemic hypoxic encephalopathy.48 Many studies are currently ongoing to determine additional evidence-based indications for mild therapeutic hypothermia.This article presented numerous possibilities and methods for the induction and maintenance of therapeutic hypothermia. There are several feasible, efficient and affordable options to implement mild therapeutic hypothermia in any intensive care setting – neither financial restraints nor infrastructural issues should therefore be reasons not to provide patients adequate neuroprotective treatment.In summary, a recommendation for any specific cooling method or any particular sedation regimen is not possible yet – more evidence and further clinical studies are needed to establish optimized hypothermia treatment procedures.

Conflict of interest

Dr. Oliver Kimberger: No conflict of interest.Dr. Andrea Kurz: No conflict of interest.
• For a fast induction and efficient maintenance of mild therapeutic hypothermia shivering and vasoconstriction thresholds have to be lowered.
• Apart from general anaesthetics meperidine, dexmedetomidine, nefopam, buspirone and skin warming lower thermoregulatory thresholds.
• With a well balanced combination of different anti-shivering drugs mild therapeutic hypothermia can be induced and maintained in awake patients, e.g. to allow neurological monitoring.
• Accurate measurement of core temperature during mild therapeutic hypothermia is very important. Pulmonary artery, oesophagus, nasopharynx, bladder or tympanic temperature should be used for core temperature measurement.
• During hypothermia the half-life of all drugs is notably prolonged, and dosing has to be adjusted accordingly.
• There are many efficient cooling methods commercially available, an evidence-based recommendation for any specific method has yet to be established.
• Research is warranted on new drugs lowering the shivering threshold without concomitant sedation and respiratory toxicity.
• Further large clinical trials are necessary to explore additional indications for mild therapeutic hypothermia.

References

1 D.I. Sessler, E.H. Rubinstein and A. Moayeri, Physiologic responses to mild perianesthetic hypothermia in humans, Anesthesiology 75 (4) (1991), pp. 594–610. View Record in Scopus | Cited By in Scopus (112)
2 J. Xiong, A. Kurz and D.I. Sessler et al., Isoflurane produces marked and nonlinear decreases in the vasoconstriction and shivering thresholds, Anesthesiology 85 (2) (1996), pp. 240–245. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (78)
3 T. Ikeda, J.S. Kim and D.I. Sessler et al., Isoflurane alters shivering patterns and reduces maximum shivering intensity, Anesthesiology 88 (4) (1998), pp. 866–873. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
4 R. Annadata, D.I. Sessler and F. Tayefeh et al., Desflurane slightly increases the sweating threshold but produces marked, nonlinear decreases in the vasoconstriction and shivering thresholds, Anesthesiology 83 (6) (1995), pp. 1205–1211. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (84)
5 T. Matsukawa, A. Kurz and D.I. Sessler et al., Propofol linearly reduces the vasoconstriction and shivering thresholds, Anesthesiology 82 (5) (1995), pp. 1169–1180. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (143)
6 A. Kurz, D.I. Sessler and R. Annadata et al., Midazolam minimally impairs thermoregulatory control, Anesthesia & Analgesia 81 (2) (1995), pp. 393–398. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (42)
7 J.M. Wheelahan, K. Leslie and B.S. Silbert, Epidural fentanyl reduces the shivering threshold during epidural lidocaine anesthesia, Anesthesia & Analgesia 87 (3) (1998), pp. 587–590. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
8 A. Kurz, J.C. Go and D.I. Sessler et al., Alfentanil slightly increases the sweating threshold and markedly reduces the vasoconstriction and shivering thresholds, Anesthesiology 83 (2) (1995), pp. 293–299. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (92)
9 P. Alfonsi, D.I. Sessler and B. Du Manoir et al., The effects of meperidine and sufentanil on the shivering threshold in postoperative patients, Anesthesiology 89 (1) (1998), pp. 43–48 pascal.alfonsi@apr.ap-hop-paris.fr. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (48)
10 A. Kurz, T. Ikeda and D.I. Sessler et al., Meperidine decreases the shivering threshold twice as much as the vasoconstriction threshold, Anesthesiology 86 (5) (1997), pp. 1046–1054. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (78)
11 A.L. Pauca, R.T. Savage, S. Simpson and R.C. Roy, Effect of pethidine, fentanyl and morphine on post-operative shivering in man, Acta Anaesthesiologica Scandinavica 28 (2) (1984), pp. 138–143. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (56)
12 M. Mokhtarani, A.N. Mahgoub and N. Morioka et al., Buspirone and meperidine synergistically reduce the shivering threshold, Anesthesia & Analgesia 93 (5) (2001), pp. 1233–1239. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (57)
13 J.E. Mackenzie and L.W. Frank, Influence of pretreatment with a monoamine oxidase inhibitor (phenelzine) on the effects of buprenorphine and pethidine in the conscious rabbit, British Journal of Anaesthesia 60 (2) (1988), pp. 216–221. View Record in Scopus | Cited By in Scopus (9)
14 K.G. Helgesen, O. Ellingsen and A. Ilebekk, Inotropic effect of meperidine: influence of receptor and ion channel blockers in the rat atrium, Anesthesia & Analgesia 70 (5) (1990), pp. 499–506. View Record in Scopus | Cited By in Scopus (5)
15 K. Kaya, A. Babacan and M. Beyazova et al., Effects of perineural opioids on nerve conduction of N. suralis in man, Acta Neurologica Scandinavica 85 (5) (1992), pp. 337–339. View Record in Scopus | Cited By in Scopus (15)
16 M. Kurz, K.G. Belani and D.I. Sessler et al., Naloxone, meperidine, and shivering, Anesthesiology 79 (6) (1993), pp. 1193–1201. View Record in Scopus | Cited By in Scopus (55)
17 L.P. Kammersgaard, B.H. Rasmussen and H.S. Jorgensen et al., Feasibility and safety of inducing modest hypothermia in awake patients with acute stroke through surface cooling: A case-control study: the Copenhagen Stroke Study, Stroke 31 (9) (2000), pp. 2251–2256. View Record in Scopus | Cited By in Scopus (130)
18 S.R. Dixon, R.J. Whitbourn and M.W. Dae et al., Induction of mild systemic hypothermia with endovascular cooling during primary percutaneous coronary intervention for acute myocardial infarction, Journal of the American College of Cardiology 40 (11) (2002), pp. 1928–1934. Article | PDF (175 K) | View Record in Scopus | Cited By in Scopus (113)
19 P.D. Lyden, R.L. Allgren and K. Ng et al., Intravascular Cooling in the Treatment of Stroke (ICTuS): Early Clinical Experience, Journal of Stroke and Cerebrovascular Diseases 14 (3) (2005), pp. 107–114. Article | PDF (232 K) | View Record in Scopus | Cited By in Scopus (20)
20 K.Z. Guluma, T.M. Hemmen and S.E. Olsen et al., A trial of therapeutic hypothermia via endovascular approach in awake patients with acute ischemic stroke: methodology, Academic Emergency Medicine 13 (8) (2006), pp. 820–827. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (33)
21 A.G. Doufas, C.M. Lin and M.I. Suleman et al., Dexmedetomidine and meperidine additively reduce the shivering threshold in humans, Stroke 34 (5) (2003), pp. 1218–1223. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (44)
22 P. Talke, F. Tayefeh and D.I. Sessler et al., Dexmedetomidine does not alter the sweating threshold, but comparably and linearly decreases the vasoconstriction and shivering thresholds, Anesthesiology 87 (4) (1997), pp. 835–841. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (65)
23 G. Nicolaou, A.A. Chen and C.E. Johnston et al., Clonidine decreases vasoconstriction and shivering thresholds, without affecting the sweating threshold, Canadian Journal of Anaesthesia 44 (6) (1997), pp. 636–642. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (21)
24 K.P. Lesch, B. Poten, K. Sohnle and H.M. Schulte, Pharmacology of the hypothermic response to 5-HT1A receptor activation in humans, European Journal of Clinical Pharmacology 39 (1) (1990), pp. 17–19. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (58)
25 H.S. Lee and H.Y. Meltzer, Buspirone does not produce a 5-HT1A-mediated decrease in temperature in man, Journal of Neural Transmission General Section 86 (1) (1991), pp. 71–76. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
26 R.C. Heel, R.N. Brogden and G.E. Pakes et al., Nefopam: a review of its pharmacological properties and therapeutic efficacy, Drugs 19 (4) (1980), pp. 249–267. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (58)
27 F. Guirimand, X. Dupont and D. Bouhassira et al., Nefopam strongly depresses the nociceptive flexion (R(III)) reflex in humans, Pain 80 (1–2) (1999), pp. 399–404. Article | PDF (162 K) | View Record in Scopus | Cited By in Scopus (36)
28 J.C. Gasser and J.W. Bellville, Respiratory effects of nefopam, Clinical Pharmacology & Therapeutics 18 (2) (1975), pp. 175–179. View Record in Scopus | Cited By in Scopus (11)
29 F. Bilotta, F. Ferri and F. Giovannini et al., Nefopam or clonidine in the pharmacologic prevention of shivering in patients undergoing conscious sedation for interventional neuroradiology, Anaesthesia 60 (2) (2005), pp. 124–128. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
30 P. Alfonsi, F. Adam and A. Passard et al., Nefopam, a nonsedative benzoxazocine analgesic, selectively reduces the shivering threshold in unanesthetized subjects, Anesthesiology 100 (1) (2004), pp. 37–43. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
31 S.N. Piper, S.W. Suttner and C.C. Schmidt et al., Nefopam and clonidine in the prevention of postanaesthetic shivering, Anaesthesia 54 (7) (1999), pp. 695–699. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (39)
32 G. Rosa, D. Dell'Utri and G. Conti et al., Efficacy of nefopam for the prevention and treatment of amphotericin B-induced shivering, Archives of Internal Medicine 157 (14) (1997), pp. 1589–1592. View Record in Scopus | Cited By in Scopus (11)
33 G. Rosa, G. Pinto and P. Orsi et al., Control of post anaesthetic shivering with nefopam hydrochloride in mildly hypothermic patients after neurosurgery, Acta Anaesthesiologica Scandinavica 39 (1) (1995), pp. 90–95. View Record in Scopus | Cited By in Scopus (43)
*34 C. Cheng, T. Matsukawa and D.I. Sessler et al., Increasing mean skin temperature linearly reduces the core-temperature thresholds for vasoconstriction and shivering in humans, Anesthesiology 82 (5) (1995), pp. 1160–1168. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (111)
35 P. Alfonsi, K.E. Nourredine and F. Adam et al., Effect of postoperative skin-surface warming on oxygen consumption and the shivering threshold, Anaesthesia 58 (12) (2003), pp. 1228–1234. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
36 O. Kimberger, S.Z. Ali and M. Markstaller et al., Meperidine and skin surface warming additively reduce the shivering threshold: a volunteer study, Critical Care 11 (1) (2007), p. R29. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)
*37 HACA-Group. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med, 346(8): 549–56.
38 O. Plattner, A. Kurz and D.I. Sessler et al., Efficacy of intraoperative cooling methods, Anesthesiology 87 (5) (1997), pp. 1089–1095. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (32)
39 D.I. Sessler and A. Moayeri, Skin-surface warming: heat flux and central temperature, Anesthesiology 73 (2) (1990), pp. 218–224. View Record in Scopus | Cited By in Scopus (78)
40 J.M. Hynson and D.I. Sessler, Intraoperative warming therapies: a comparison of three devices, Journal of Clinical Anesthesia 4 (3) (1992), pp. 194–199. Abstract | View Record in Scopus | Cited By in Scopus (121)
41 M.A. Theard, R. Tempelhoff and C.M. Crowder et al., Convection versus conduction cooling for induction of mild hypothermia during neurovascular procedures in adults, Journal of Neurosurgical Anesthesiology 9 (3) (1997), pp. 250–255. View Record in Scopus | Cited By in Scopus (17)
42 S.A. Mayer, R.G. Kowalski and M. Presciutti et al., Clinical trial of a novel surface cooling system for fever control in neurocritical care patients, Critical Care Medicine 32 (12) (2004), pp. 2508–2515. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (70)
43 M. Haugk, F. Sterz and M. Grassberger et al., Feasibility and efficacy of a new non-invasive surface cooling device in post-resuscitation intensive care medicine, Resuscitation 75 (1) (2007), pp. 76–81. Article | PDF (199 K) | View Record in Scopus | Cited By in Scopus (23)
44 N. Nesher, G. Uretzky and S. Insler et al., Thermo-wrap technology preserves normothermia better than routine thermal care in patients undergoing off-pump coronary artery bypass and is associated with lower immune response and lesser myocardial damage, The Journal of Thoracic and Cardiovascular Surgery 129 (6) (2005), pp. 1371–1378. Abstract | View Record in Scopus | Cited By in Scopus (8)
45 C.K. Hofer, M. Worn and R. Tavakoli et al., Influence of body core temperature on blood loss and transfusion requirements during off-pump coronary artery bypass grafting: a comparison of 3 warming systems, The Journal of Thoracic and Cardiovascular Surgery 129 (4) (2005), pp. 838–843. Abstract | View Record in Scopus | Cited By in Scopus (17)
46 M.R. Battin, J. Penrice, T.R. Gunn and A.J. Gunn, Treatment of term infants with head cooling and mild systemic hypothermia (35.0 degrees C and 34.5 degrees C) after perinatal asphyxia, Pediatrics 111 (2) (2003), pp. 244–251. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (93)
47 W. Qiu, H. Shen and Y. Zhang et al., Noninvasive selective brain cooling by head and neck cooling is protective in severe traumatic brain injury, Journal of Clinical Neuroscience 13 (10) (2006), pp. 995–1000. Article | PDF (139 K) | View Record in Scopus | Cited By in Scopus (10)
*48 P.D. Gluckman, J.S. Wyatt and D. Azzopardi et al., Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: multicentre randomised trial, Lancet 365 (9460) (2005), pp. 663–670. Article | PDF (120 K) | View Record in Scopus | Cited By in Scopus (368)
49 S.O. Bello, Selective head cooling after neonatal encephalopathy, Lancet 365 (9471) (2005), p. 1619. Article | PDF (60 K)
50 A.R. Horn, D.L. Woods and C. Thompson et al., Selective cerebral hypothermia for post-hypoxic neuroprotection in neonates using a solid ice cap, South African Medical Journal 96 (9 Pt 2) (2006), pp. 976–981. View Record in Scopus | Cited By in Scopus (10)
51 A. Rajek, R. Greif and D.I. Sessler et al., Core cooling by central venous infusion of ice-cold (4 degrees C and 20 degrees C) fluid: isolation of core and peripheral thermal compartments, Anesthesiology 93 (3) (2000), pp. 629–637. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (67)
52 S. Bernard, M. Buist, O. Monteiro and K. Smith, Induced hypothermia using large volume, ice-cold intravenous fluid in comatose survivors of out-of-hospital cardiac arrest: a preliminary report, Resuscitation 56 (1) (2003), pp. 9–13. Article | PDF (93 K) | View Record in Scopus | Cited By in Scopus (183)
53 I. Virkkunen, A. Yli-Hankala and T. Silfvast, Induction of therapeutic hypothermia after cardiac arrest in prehospital patients using ice-cold Ringer's solution: a pilot study, Resuscitation 62 (3) (2004), pp. 299–302. Article | PDF (57 K) | View Record in Scopus | Cited By in Scopus (58)
54 A. Kliegel, H. Losert and F. Sterz et al., Cold simple intravenous infusions preceding special endovascular cooling for faster induction of mild hypothermia after cardiac arrest–a feasibility study, Resuscitation 64 (3) (2005), pp. 347–351. Article | PDF (86 K) | View Record in Scopus | Cited By in Scopus (52)
*55 K.H. Polderman, E.R. Rijnsburger, S.M. Peerdeman and A.R. Girbes, Induction of hypothermia in patients with various types of neurologic injury with use of large volumes of ice-cold intravenous fluid, Critical Care Medicine 33 (12) (2005), pp. 2744–2751. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (42)
56 A. Kliegel, A. Janata and C. Wandaller et al., Cold infusions alone are effective for induction of therapeutic hypothermia but do not keep patients cool after cardiac arrest, Resuscitation 73 (1) (2007), pp. 46–53. Article | PDF (169 K) | View Record in Scopus | Cited By in Scopus (29)
57 E. Schmutzhard, K. Engelhardt and R. Beer et al., Safety and efficacy of a novel intravascular cooling device to control body temperature in neurologic intensive care patients: a prospective pilot study, Critical Care Medicine 30 (11) (2002), pp. 2481–2488. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (67)
58 K.H. Polderman and J. Callaghan, Equipment review: cooling catheters to induce therapeutic hypothermia?, Critical Care 10 (6) (2006), p. 234. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)
59 L. Covaciu, M. Allers and P. Enblad et al., Intranasal selective brain cooling in pigs, Resuscitation 76 (1) (January 2008), pp. 83–88.
60 M.M. Zviman, A. Roguin and A. Jacobs et al., A new method for inducing hypothermia during cardiac arrest, Critical Care Medicine 32 (9 Suppl.) (2004), pp. S369–S373. Full Text via CrossRef
61 P.J. Safar and P.M. Kochanek, Therapeutic hypothermia after cardiac arrest, The New England Journal of Medicine 346 (8) (2002), pp. 612–613. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (95)
62 Y.S. Wen, M.S. Huang, M.T. Lin and C.H. Lee, Rapid brain cooling by hypothermic retrograde jugular vein flush, The Journal of Trauma 58 (3) (2005), pp. 577–581. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
63 S.S. Yang, M.J. Jeng and R. McShane et al., Cold perfluorochemical-induced hypothermia protects lung integrity in normal rabbits, Biology of the Neonate 87 (1) (2005), pp. 60–65. Article | PDF (231 K) | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
64 K. Mori, Y. Itoh and J. Saito et al., Post-resuscitative hypothermic bypass reduces ischemic brain injury in swine, Academic Emergency Medicine 8 (10) (2001), pp. 937–945. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
65 K. Mori, J. Saito and Y. Kurata et al., Rapid development of brain hypothermia using femoral–carotid bypass, Academic Emergency Medicine 8 (4) (2001), pp. 303–308. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)
66 M. Kawaguchi, S. Inoue and T. Sakamoto et al., The effects of prostaglandin E1 on intraoperative temperature changes and the incidence of postoperative shivering during deliberate mild hypothermia for neurosurgical procedures, Anesthesia & Analgesia 88 (2) (1999), pp. 446–451. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)
67 S. Inoue, M. Kawaguchi and T. Sakamoto et al., Amrinone can accelerate the cooling rate of core temperature during deliberate mild hypothermia for neurosurgical procedures, British Journal of Anaesthesia 86 (5) (2001), pp. 663–668. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
68 S. Inoue, M. Kawaguchi and T. Sakamoto et al., High-dose amrinone is required to accelerate rewarming from deliberate mild intraoperative hypothermia for neurosurgical procedures, Anesthesiology 97 (1) (2002), pp. 116–123. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
69 K. Hebbar, J.D. Fortenberry and K. Rogers et al., Comparison of temporal artery thermometer to standard temperature measurements in pediatric intensive care unit patients, Pediatric Critical Care Medicine 6 (5) (2005), pp. 557–561. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
70 M.J. Kresch, Axillary temperature as a screening test for fever in children, The Journal of Pediatrics 104 (4) (1984), pp. 596–599. Abstract | View Record in Scopus | Cited By in Scopus (25)
71 J.Y. Lefrant, L. Muller and J.E. de La Coussaye et al., Temperature measurement in intensive care patients: comparison of urinary bladder, oesophageal, rectal, axillary, and inguinal methods versus pulmonary artery core method, Intensive Care Medicine 29 (3) (2003), pp. 414–418. View Record in Scopus | Cited By in Scopus (39)
72 M.B. Jean-Mary, J. Dicanzio, J. Shaw and H.H. Bernstein, Limited accuracy and reliability of infrared axillary and aural thermometers in a pediatric outpatient population, The Journal of pediatrics 141 (5) (2002), pp. 671–676. Abstract | View Record in Scopus | Cited By in Scopus (16)
73 M. Imamura, T. Matsukawa and M. Ozaki et al., The accuracy and precision of four infrared aural canal thermometers during cardiac surgery, Acta Anaesthesiologica Scandinavica 42 (10) (1998), pp. 1222–1226. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (21)
74 A. Petersen-Smith, N. Barber and D.K. Coody et al., Comparison of aural infrared with traditional rectal temperatures in children from birth to age three years, The Journal of Pediatrics 125 (1) (1994), pp. 83–85. Abstract | View Record in Scopus | Cited By in Scopus (30)
75 F.A. Rhoads and J. Grandner, Assessment of an aural infrared sensor for body temperature measurement in children, Clinical Pediatrics 29 (2) (1990), pp. 112–115. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
76 C.G. Cattaneo, S.M. Frank and T.W. Hesel et al., The accuracy and precision of body temperature monitoring methods during regional and general anesthesia, Anesthesia & Analgesia 90 (4) (2000), pp. 938–945. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (32)
77 D. Callanan, Detecting fever in young infants: reliability of perceived, pacifier, and temporal artery temperatures in infants younger than 3 months of age, Pediatric Emergency Care 19 (4) (2003), pp. 240–243. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)
78 M.I. Suleman, A.G. Doufas and O. Akca et al., Insufficiency in a new temporal-artery thermometer for adult and pediatric patients, Anesthesia & Analgesia 95 (1) (2002), pp. 67–71 [table of contents]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
79 O. Kimberger, D. Cohen, U. Illievich and R. Lenhardt, Temporal artery versus bladder thermometry during perioperative and intensive care unit monitoring, Anesthesia & Analgesia 105 (4) (2007), pp. 1042–1047 [table of contents]. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
*80 J.L. Moran, J.V. Peter and P.J. Solomon et al., Tympanic temperature measurements: are they reliable in the critically ill? A clinical study of measures of agreement, Critical Care Medicine 35 (1) (2007), pp. 155–164. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
81 D.S. Greenes and G.R. Fleisher, When body temperature changes, does rectal temperature lag?, The Journal of Pediatrics 144 (6) (2004), pp. 824–826. Abstract | View Record in Scopus | Cited By in Scopus (15)
82 L. Aziz, K. Ono and Y. Ohta et al., Effect of hypothermia on the in vitro potencies of neuromuscular blocking agents and on their antagonism by neostigmine, British Journal of Anaesthesia 73 (5) (1994), pp. 662–666. View Record in Scopus | Cited By in Scopus (6)
83 N.J. Smeulers, J.M. Wierda and L. van den Broek et al., Hypothermic cardiopulmonary bypass influences the concentration–response relationship and the biodisposition of rocuronium, European Journal of Anaesthesiology Supplement 11 (1995), pp. 91–94. View Record in Scopus | Cited By in Scopus (7)
84 G. Cammu, J. Coddens, J. Hendrickx and T. Deloof, Dose requirements of infusions of cisatracurium or rocuronium during hypothermic cardiopulmonary bypass, British Journal of Anaesthesia 84 (5) (2000), pp. 587–590. View Record in Scopus | Cited By in Scopus (12)
85 A.J. England, X. Wu and K.M. Richards et al., The influence of cold on the recovery of three neuromuscular blocking agents in man, Anaesthesia 51 (3) (1996), pp. 236–240. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
86 H.G. Fritz, M. Holzmayr and B. Walter et al., The effect of mild hypothermia on plasma fentanyl concentration and biotransformation in juvenile pigs, Anesthesia & Analgesia 100 (4) (2005), pp. 996–1002. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
*87 M.A. Tortorici, P.M. Kochanek and S.M. Poloyac, Effects of hypothermia on drug disposition, metabolism, and response: A focus of hypothermia-mediated alterations on the cytochrome P450 enzyme system, Critical Care Medicine 35 (9) (2007), pp. 2196–2204. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
88 S.A. Bernard and M. Buist, Induced hypothermia in critical care medicine: a review, Critical Care Medicine 31 (7) (2003), pp. 2041–2051. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (112)
89 H. Schmied, A. Kurz and D.I. Sessler et al., Mild hypothermia increases blood loss and transfusion requirements during total hip arthroplasty, Lancet 347 (8997) (1996), pp. 289–292. Article | PDF (639 K) | View Record in Scopus | Cited By in Scopus (345)
90 M. Winkler, O. Akca and B. Birkenberg et al., Aggressive warming reduces blood loss during hip arthroplasty, Anesthesia & Analgesia 91 (4) (2000), pp. 978–984. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (61)
*91 A. Kurz, D.I. Sessler and R. Lenhardt, Perioperative normothermia to reduce the incidence of surgical-wound infection and shorten hospitalization. Study of Wound Infection and Temperature Group, The New England Journal of Medicine 334 (19) (1996), pp. 1209–1215. View Record in Scopus | Cited By in Scopus (807)
92 H.W. Hopf, T.K. Hunt and J.M. West et al., Wound tissue oxygen tension predicts the risk of wound infection in surgical patients, Archives of Surgery 132 (9) (1997), pp. 997–1004 [discussion 1005].
93 B. Beilin, Y. Shavit and J. Razumovsky et al., Effects of mild perioperative hypothermia on cellular immune responses, Anesthesiology 89 (5) (1998), pp. 1133–1140. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (91)
94 C. Wenisch, E. Narzt and D.I. Sessler et al., Mild intraoperative hypothermia reduces production of reactive oxygen intermediates by polymorphonuclear leukocytes, Anesthesia & Analgesia 82 (4) (1996), pp. 810–816. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (88)
95 S.M. Frank, L.A. Fleisher and M.J. Breslow et al., Perioperative maintenance of normothermia reduces the incidence of morbid cardiac events. A randomized clinical trial, The Journal of the American Medical Association 277 (14) (1997), pp. 1127–1134. View Record in Scopus | Cited By in Scopus (427)
96 J.P. Nolan, P.T. Morley, T.L. Hoek and R.W. Hickey, Therapeutic hypothermia after cardiac arrest. An advisory statement by the Advancement Life support Task Force of the International Liaison committee on Resuscitation, Resuscitation 57 (3) (2003), pp. 231–235. Abstract | PDF (177 K) | View Record in Scopus | Cited By in Scopus (162)
*97 S.A. Bernard, T.W. Gray and M.D. Buist et al., Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia, The New England Journal of Medicine 346 (8) (2002), pp. 557–563. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1072)
  相似文献   

5.
Haemostatic side-effects may be either beneficial or unwanted depending on the status of a given patient. Attenuation of hypercoagulability may be beneficial in patients at risk for thrombotic episodes. In patients at risk for bleeding, however, the anticoagulant effects may translate into blood loss and transfusion requirements. Judging by currently available evidence, adverse haemostasis effects appear to be inherent to colloidal molecules, although dextran, hetastarch and pentastarch definitely have a more pronounced impact than tetrastarch, gelatin and albumin (evidence level Ib). Especially in the era of colloids with minimal, predictable and well-described influences on the haemostatic system, other adverse side-effects become of utmost importance in decision making in fluid management. Tetrastarch appears to be a suitable volume expander in the routine intensive care setting due to its high volume efficacy and safety index. Contra-indications and maximum daily doses need to be acknowledged. Gelatins have even lower inhibitory effects on clot strength compared with tetrastarch, but volume efficacy is also lower and anaphylactoid reactions are more frequent. Dextrans are potent anticoagulants with a high risk for adverse reactions. Albumin has negligible effects on haemostasis, but low volume efficacy and costs limit the use of a blood product as a routine volume replacement fluid.
• colloidal molecules inhibit platelet function (in carrier solutions without calcium): dextran ≥ hetastarch > pentastarch > gelatin ≥ tetrastarch, albumin
• colloidal molecules decrease vWF and factor VIII: dextran ≥ hetastarch > pentastarch > gelatin ≥ tetrastarch, albumin
• colloidal molecules induce hypocoagulability and decrease fibrin polymerization: dextran > hetastarch > pentastarch > tetrastarch > gelatin > albumin
• to avoid potential acidosis-induced changes in haemostasis, plasma-adapted carrier solutions may be used instead of saline-based solutions
• effect of colloidal molecules on blood loss: dextran > hetastarch > pentastarch > gelatin, tetrastarch, albumin
• postoperative fluid therapy (in the absence of bleeding complications) should improve rheology and should not aggravate postoperative activation of coagulation. A moderate inhibitory effect on thrombin generation and platelet hyper-reactivity may be favourable
• crystalloids, HES and gelatins are most frequently used in postoperative hypercoagulability
• fluids with anticoagulant and antiplatelet side-effects (such as dextrans) attenuate hypercoagulability[50] and [94], but cannot be used as a substitute for thrombosis prophylaxis if indicated
• there is no clear recommendation for any particular fluid therapy and for different types of colloids in sepsis
• tetrastarch, gelatins and crystalloids are most frequently used in sepsis-associated coagulopathy; tetrastarch may exert anti-inflammatory effects while preserving haemostasis. Gelatins have lower inhibitory effects on clot strength compared with tetrastarch, but their volume efficacy is also lower. Albumin has negligible effects on haemostasis, but low volume efficacy and costs limit the use of a blood product as a routine volume replacement fluid in sepsis. Dextrans are not recommended in sepsis due to potent anticoagulant effects with a high risk for adverse reactions
• in patients with severe liver failure, fluids with negligible effects on haemostasis and organ function are required. Potato-derived tetrastarch was associated with mild-to-moderate hyperbilirubinaemia61 and is contraindicated in patients with severe hepatic impairment
• long-term use of hyperoncotic solutions is not indicated in intensive care patients at risk for renal failure
• there are differences between older generations of HES and tetrastarch. Hetastarches and pentastarches should not be used in renal failure
• contra-indications (e.g. anuria) and maximum daily doses for tetrastarch need to be acknowledged
• binding of organic compounds such as prostaglandins to amylopectin and linear chains of amylose in potato-derived HES preparations may impair the antiplatelet effect97
• the content of calcium in the crystalloid carrier solutions is unlikely to affect calcium re-infusion after the haemofilter in citrate anticoagulation
• in the absence of firm evidence98, tetrastarch appears to be superior to other fluids in massive bleeding due to its volume efficacy and safety
• small-volume resuscitation is reported to result in smaller haemostatic derangements compared with conventional colloidal resuscitation.81
• it remains to be determined whether extracellular coating impairs platelet procoagulant activity by modifying the binding of constituents of the prothrombinase and tenase complex to the negatively charged phospholipids exposed on activated platelets
• the exact mechanisms for the decrease in factor VIII and vWF by colloids has not been elucidated fully
• studies are warranted to elucidate the effect of potato-derived tetrastarch on haemostasis, especially after high-dose, long-term application
• future papers on HES need to clearly indicate molar substitution, C2/C6 ratio, molecular weight, suspension medium and oncotic characteristics in order to permit precise interpretation of the results
• large-scale clinical studies on the clinical relevance of acid-base disturbance after colloid infusions are warranted
• a head-to-head comparison of balanced tetrastarch and balanced gelatins on blood loss and transfusion requirements in patients at risk for bleeding is needed
• the relationship between bleeding and laboratory coagulation parameters (e.g. clot polymerization) in the presence of synthetic colloids remains open to question in humans
• a head-to-head comparison of balanced tetrastarch, balanced gelatins and albumin on side-effects on haemostasis in septic patients is needed
• adequately powered in-vivo studies employing a clinically relevant methodology are needed. Infusion of 20 mL/kg colloid within 10 min does not necessarily represent common clinical situations
• the effect of fluid therapy on micro-embolization, organ and cognitive functions warrants evaluation in well-designed experimental settings
• although often used as a parameter of effectiveness99, mortality may not be a suitable outcome parameter due to the many confounding factors, even in well-standardized animal experiments.100 Survival rate was comparable in rabbits resuscitated with gelatin, HES or crystalloid, but survival time was longest in HES-treated animals39

References

*1 S. Kozek-Langenecker, Effects of hydroxyethyl starch solutions on hemostasis, Anesthesiology 103 (2005), pp. 654–660. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (49)
2 S. Kozek-Langenecker and G. Scharbert, Effect of hydroxyethyl starches on hemostasis, Transfusion Alternatives in Transfusion Medicine 9 (2007), pp. 173–181. Full Text via CrossRef
3 P. Van der Linden and B. Ickx, The effects of colloid solutions on hemostasis, Canadian Journal of Anaesthesia 53 (2006), pp. S30–S39. View Record in Scopus | Cited By in Scopus (14)
4 B. Stögermüller, J. Stark and H. Willschke et al., The effects of hydroxyethyl starch 200 kD on platelet function, Anesthesia and Analgesia 91 (2000), pp. 823–827. View Record in Scopus | Cited By in Scopus (50)
5 A. Franz, P. Bräunlich and T. Gamsjäger et al., The effects of hydroxyethyl starches of varying molecular weight on platelet function, Anesthesia and Analgesia 92 (2001), pp. 1402–1407. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (81)
6 I. Huttner, J. Boldt and G. Haisch et al., Influence of different colloids on molecular markers of haemostasis and platelet function in patients undergoing major abdominal surgery, British Journal of Anaesthesia 85 (2000), pp. 417–423. View Record in Scopus | Cited By in Scopus (30)
7 M. Omar, T. Shouk and M. Khaleq, Activity of blood coagulation and fibrinolysis during and after hydroxyethyl starch (HES) colloidal volume replacement, Clinical Biochemistry 32 (1999), pp. 269–274. Article | PDF (165 K) | View Record in Scopus | Cited By in Scopus (14)
8 T. Gamsjäger, B. Gustorff and S. Kozek-Langenecker, The effects of hydroxyethyl starches on intracellular calcium in platelets, Anesthesia and Analgesia 95 (2002), pp. 866–869. View Record in Scopus | Cited By in Scopus (13)
9 E. Deusch, T. Gamsjäger and H. Kress et al., Binding of hydroxyethyl starch molecules to the platelet surface, Anesthesia and Analgesia 97 (2003), pp. 680–683. View Record in Scopus | Cited By in Scopus (26)
10 E. deJonge and M. Levi, Effects of different plasma substitutes on blood coagulation: a comparative review, Critical Care Medicine 29 (2001), pp. 1261–1267. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (106)
11 U. Thaler, E. Deusch and S. Kozek-Langenecker, In vitro effects of various gelatin solutions on platelet function and their comparison to hydroxyethyl starch solutions, Anaesthesia 60 (2005), pp. 554–559. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (16)
12 E. deJonge, M. Levi and F. Berends et al., Impaired haemostasis by intravenous administration of a gelatin-based plasma expander in human subjects, Thrombosis and Haemostasis 79 (1998), pp. 286–290. View Record in Scopus | Cited By in Scopus (46)
*13 B. Kheirabadi, J. Crissey and R. Deguzman et al., Effects of synthetic versus natural colloid resuscitation on inducing dilutional coagulopathy and increasing hemorrhage in rabbits, The Journal of Trauma 64 (2008), pp. 1218–1228. View Record in Scopus | Cited By in Scopus (2)
14 J. Battle, Dr F and M. Lopez Fernandez et al., Effect of dextran on factor VIII/von Willebrand factor structure and function, Thrombosis and Haemostasis 54 (1985), pp. 697–699.
15 M. Mittermayr, W. Streif and T. Haas et al., Haemostatic changes after crystalloid or colloid fluid administration during orthopedic surgery: the role of fibrinogen administration, Anesthesia and Analgesia 105 (2007), pp. 905–917. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
16 G. Petroianu, W. Maleck and K. Koetter et al., Effect of in vitro hemodilution with hydroxyethyl starch and dextran on the activity of plasma clotting factors, Critical Care Medicine 31 (2003), pp. 250–254. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
17 E. Entholzer, L. Mielke and A. Calatzis et al., Coagulation effects of a recently developed hydroxyethyl starch (HES 130/0.4) compared to hydroxyethyl starches with higher molecular weight, Acta Anaesthesiologica Scandinavica 44 (2000), pp. 1116–1121.
18 M. Tobias, D. Wambold and M. Pilla et al., Differential effects of serial hemodilution with hydroxyethyl starch, albumin, and 0.9% saline on whole blood coagulation, Journal of Clinical Anesthesia 10 (1998), pp. 366–371. Article | PDF (279 K) | View Record in Scopus | Cited By in Scopus (33)
19 D. Fries, P. Innerhofer and A. Klingler et al., The effects of the combined administration of colloids and lactated Ringer's solution on the coagulation system: an in vitro study using thrombelastograph coagulation analysis (ROTEG), Anesthesia and Analgesia 94 (2002), pp. 1280–1287. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (72)
20 V. Nielsen, Effects of PentaLyte and Voluven hemodilution on plasma coagulation kinetics in the rabbit: role of thrombin-fibrinogen and factor XIII-fibrin polymer interactions, Acta Anaesthesiologica Scandinavica 49 (2005), pp. 1263–1271. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (17)
21 R. Strauss, B. Pennell and D. Stump, A randomized controlled trial comparing the hemostatic effects of pentastarch versus hetastarch, Transfusion 42 (2002), pp. 27–36. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (37)
22 J. Boldt, G. Haisch and S. Suttner et al., Effects of a new modified, balanced hydroxyethyl starch preparation (Hextend) on measures of coagulation, British Journal of Anaesthesia 89 (2002), pp. 722–728. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (56)
23 V. Nielsen, Hemodilution modulates the time of onset and rate of fibrinolysis in human and rabbit plasma, The Journal of Heart and Lung Transplantation 25 (2006), pp. 1344–1352. Abstract | View Record in Scopus | Cited By in Scopus (11)
24 T. Niemi and A. Kuitunen, Artificial colloids impair haemostasis. An in vitro study using thromboelastometry coagulation analysis, Acta Anaesthesiologica Scandinavica 49 (2005), pp. 373–378. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
25 E. deJonge, M. Levi and R. Buller et al., Decreased circulating levels of von Willebrand factor after intravenous administration of a rapidly degradable hydroxyethyl starch (HES 200/0.5/6) in healthy human subjects, Intensive Care Medicine 27 (2001), pp. 1825–1829. View Record in Scopus | Cited By in Scopus (21)
26 M. Jamnicki, T. Bombeli and B. Seifert et al., Low- and medium-molecular-weight hydroxyethyl starches: comparison of their effect on blood coagulation, Anesthesiology 93 (2000), pp. 1231–1237. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (52)
27 S. Kapiotis, P. Quehenberger and H. Eichler et al., Effect of hydroxyethyl starch on the activity of blood coagulation and fibrinolysis in healthy volunteers: comparison with albumin, Critical Care Medicine 22 (1994), pp. 606–612. View Record in Scopus | Cited By in Scopus (66)
28 J. Conroy, R. Fishman and S. Reeves et al., The effects of desmopressin and 6% hydroxyethyl starch on factor VIII: C, Anesthesia and Analgesia 83 (1996), pp. 804–807. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (43)
29 J. Treib, A. Haass and G. Pindur et al., All medium starches are not the same: influence of the degree of hydroxyethyl substitution of hydroxyethyl starch on plasma volume, hemorrheologic conditions, and coagulation, Transfusion 36 (1996), pp. 450–455. View Record in Scopus | Cited By in Scopus (104)
30 C. Jungheinrich, W. Sauermann and F. Bepperling et al., Volume efficacy and reduced influence on measures of coagulation using hydroxyethyl starch 130/0.4 (6%) with an optimised in vivo molecular weight in orthopedic surgery: a randomised, double-blind study, Drugs in R&D 5 (2004), pp. 1–9. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
31 T. Neff, M. Doelberg and C. Jungheinrich et al., Repetitive large-dose infusion of the novel hydroxyethyl starch 130/0.4 in patients with severe head injury, Anesthesia and Analgesia 96 (2003), pp. 1453–1459. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (54)
32 S. Kasper, P. Meinert and S. Kampe et al., Large-dose hydroxyethyl starch 130/0.4 does not increase blood loss and transfusion requirements in coronary artery bypass surgery compared with hydroxyethyl starch 200/0.5 at recommended doses, Anesthesiology 99 (2003), pp. 42–47. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (33)
33 A. Kuitunen, M. Hynynen and E. Vahtera et al., Hydroxyethyl starch as a priming solution for cardiopulmonary bypass impairs hemostasis after cardiac surgery, Anesthesia and Analgesia 98 (2004), pp. 291–297. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
34 R. Gallandat-Huet, A. Siemons and D. Baus et al., A novel hydroxyethyl starch (Voluven) for effective perioperative plasma volume substitution in cardiac surgery, Canadian Journal of Anaesthesia 47 (2000), pp. 1207–1215. Full Text via CrossRef
35 O. Langeron, M. Doelberg and E. Ang et al., Voluven, a lower substituted novel hydroxyethyl starch (HES 130/0.4), causes fewer effects on coagulation in major orthopedic surgery than HES 200/0.5, Anesthesia and Analgesia 92 (2001), pp. 855–862. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (88)
36 B. Ellger, J. Freyhoff and H. van Aken et al., High-dose volume replacement using HES 130/0.4 during major surgery, Nederlands Tijdschrift Voor Gerontologie 19 (2006), pp. 63–68. View Record in Scopus | Cited By in Scopus (2)
37 T.G. Ruttmann, M.F. James and J.F. Viljoen, Haemodilution induces a hypercoagulable state, British Journal of Anaesthesia 76 (1996), pp. 412–414. View Record in Scopus | Cited By in Scopus (107)
38 A. McCammon, J. Wright and M. Figueroa et al., Hemodilution with albumin, but not hextend®, results in hypercoagulability as assessed by thrombelastography® in rabbits: role of heparin-dependent serpins and factor VIII complex, Anesthesia and Analgesia 95 (2002), pp. 844–850. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
*39 L. Li and X. Xu, Effect of acute progressive normovolemic hemodilution with lactated Ringer's, gelatin and hydroxyethyl starch on coagulation and survival rate in rabbits, Beijing Da Xue Xue Bao 40 (2008), pp. 292–300.
40 V. Kretschmer, A. Daraktchiev and S. Bade et al., Does hemodilution enhance coagulability?, Anästhesiologie, Intensivmedizin, Notfallmedizin, Schmerztherapie : AINS 39 (2004), pp. 751–756. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
41 P. Innerhofer, D. Fries and J. Margreiter et al., The effects of perioperatively administered colloids and crystalloids on primary platelet-mediated hemostasis and clot formation, Anesthesia and Analgesia 95 (2002), pp. 858–865. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (76)
42 M. Jamnicki, A. Zollinger and B. Seifert et al., Compromised blood coagulation: an in vitro comparison of hydroxyethyl starch 130/0.4 and hydroxyethyl starch 200/0.5 using thrombelastography, Anesthesia and Analgesia 87 (1998), pp. 989–993. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (54)
43 I. von Roten, C. Madjdpour and P. Frascarolo et al., Molar substitution and C2/C6 ratio of hydroxyethyl starch: influence on blood coagulation, British Journal of Anaesthesia 96 (2006), pp. 455–463. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)
44 C. Madjdpour, N. Dettori and P. Frascarolo et al., Molecular weight of hydroxyethyl starch: is there an effect on blood coagulation and pharmacokinetics?, British Journal of Anaesthesia 94 (2005), pp. 569–576. View Record in Scopus | Cited By in Scopus (21)
45 C. Thyes, C. Madjdpour and P. Frascarolo et al., Effect of high- and low-molecular-weight low-substituted hydroxyethyl starch on blood coagulation during acute normovolemic hemodilution in pigs, Anesthesiology 105 (2006), pp. 1228–1237. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
46 R. Strauss, D. Stump and R. Henriksen et al., Effects of hydroxyethyl starch on fibrinogen, fibrin clot formation and fibrinolysis, Transfusion 25 (1985), pp. 230–234. View Record in Scopus | Cited By in Scopus (39)
47 C. Fenger-Eriksen, J. Heslop and E. Anker-Moller et al., Thrombelastographic whole blood clot formation following ex vivo addition of plasma substitutes: improvements of the induced coagulopathy with fibrinogen concentrate, British Journal of Anaesthesia 94 (2005), pp. 324–329. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)
48 V. Nielsen, Colloids decrease clot propagation and strength: role of factor XIII-fibrin polymer and thrombin-fibrinogen interactions, Acta Anaesthesiologica Scandinavica 49 (2005), pp. 1163–1171. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (32)
49 V. Nielsen and J. Kirklin, Hydroxyethyl starch enhances argatroban-mediated decrease in clot propagation and strength by diminishing thrombin-fibrinogen interactions, Blood Coagulation & Fibrinolysis 18 (2007), pp. 49–54. View Record in Scopus | Cited By in Scopus (4)
50 S. Jones, C. Whitten and G. Despotis et al., The influence of crystalloid and colloid replacement solutions in acute normovolemic hemodilution: a preliminary survey of hemostatic markers, Anesthesia and Analgesia 96 (2003), pp. 363–368. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (30)
51 C. Konrad, T. Markl and G. Schuepfer et al., The effects of in vitro hemodilution with gelatin, hydroxyethyl starch, and lactated Ringer's solution on markers of coagulation: an analysis using Sonoclot™, Anesthesia and Analgesia 88 (1999), pp. 483–488. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)
52 T. Haas, A. Preinreich and E. Oswald et al., Effects of albumin 5% and artificial colloids on clot formation in small infants, Anaesthesia 62 (2008), pp. 1000–1007.
53 B. Ickx and P. Van der Linden, Interactions entre les solutes colloides et l'hemostase, Sang Thrombose Vaisseaux 14 (2002), pp. 408–416. View Record in Scopus | Cited By in Scopus (2)
54 M. Mittermayr, W. Streif and T. Haas et al., Effects of colloid and crystalloid solutions on endogenous activation of fibrinolysis and resistance of polymerized fibrin to recombinant tissue plasminogen activator added ex vivo, British Journal of Anaesthesia 100 (2008), pp. 307–314. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
55 M. Eriksson and T. Saldeen, Effect of dextran on plasma tissue plasminogen activator (t-PA) and plasminogen activator inhibitor-1 (PAI-1) during surgery, Acta Anaesthesiologica Scandinavica 39 (1995), pp. 163–166. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
56 H. Mohri, Acquired von Willebrand syndrome: features and management, American Journal of Hematology 81 (2006), pp. 616–623. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
57 B. Sorensen, C. Fenger-Eriksen and J. Ingerslev, Recombinant factor VIIa fails to correct coagulopathy induced by haemodilution with colloids, British Journal of Anaesthesia 94 (2005), pp. 862–863. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
58 D. Fries, A. Krismer and A. Klingler et al., Effect of fibrinogen on reversal of dilutional coagulopathy: a porcine model, British Journal of Anaesthesia 95 (2005), pp. 172–177. View Record in Scopus | Cited By in Scopus (45)
*59 T. Haas, D. Fries and C. Velik-Salchner et al., The in vitro effects of fibrinogen concentrate, factor XIII and fresh frozen plasma on impaired clot formation after 60% dilution, Anesthesia and Analgesia 106 (2008), pp. 1360–1365. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
60 G. Lehmann, G. Marx and H. Foerster, Bioequivalence comparison between hydroxyethyl starch 130/0.42/6: 1 and hydroxyethyl starch 130/0.4/9: 1, Journal of Drugs in R&D 8 (2007), pp. 229–240. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
61 O. Sander, K. Reinhart and A. Meier-Hellmann, Equivalence of hydroxyethyl starch HES 130/0.4 and HES 200/0.5 for perioperative volume replacement in mayor gynaecological surgery, Acta Anaesthesiologica Scandinavica 47 (2003), pp. 1151–1158. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (21)
62 J. Waters, A. Gottlieb and P. Schoenwald et al., Normal saline versus lactated Ringer's solution for intraoperative fluid management in patients undergoing abdominal aortic aneurysma repair: an outcome study, Anesthesia and Analgesia 93 (2001), pp. 817–822. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (71)
63 R. Stephens and M. Mythen, Optimizing intraoperative fluid therapy, Current Opinion in Anaesthesiology 16 (2003), pp. 385–392. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
64 N. Wilkes, R. Woolf and M. Mutch et al., The effects of balanced versus saline-based hetastarch and crystalloid solutions on acid-base and electrolyte status and gastric mucosal perfusion in elderly surgical patients, Anesthesia and Analgesia 93 (2001), pp. 811–816. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (121)
65 E. Base, T. Standl and A. Lassnig et al., Comparison of 6% HES 130/0.4 in a balanced electrolyte solution versus 6% HES 130/0.4 in saline solution in cardiac surgery, Critical Care 10 (2006), p. P176. Full Text via CrossRef
66 M. Engstrom, U. Schott and B. Romner et al., Acidosis impairs the coagulation: a thromboelastographic study, The Journal of Trauma 61 (2006), pp. 624–628. View Record in Scopus | Cited By in Scopus (23)
67 A. Roche, M. James and B.-G. E et al., A head-to-head comparison of the in vitro coagulation effects of saline-based and balanced electrolyte crystalloid and colloid intravenous fluids, Anesthesia and Analgesia 102 (2006), pp. 1274–1279. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
68 J. Boldt, M. Wolf and A. Mengistu, A new plasma-adapted hydroxyethylstarch preparation: in vitro coagulation studies using thrombelastography and whole blood aggregometry, Anesthesia and Analgesia 104 (2007), pp. 425–430. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (24)
69 D. Weeks, J. Jahr and J. Lim et al., Does hextend impair coagulation compared to 6% hetastarch? An ex vivo thrombelastography study, American Journal of Therapeutics 15 (2008), pp. 225–230. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
70 G. Scharbert and S. Kozek-Langenecker, Limitations of in vitro models, Anesthesia and Analgesia 105 (2007), p. 885. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
71 E. Deusch, U. Thaler and S. Kozek-Langenecker, The effects of high molecular weight hydroxyethyl starch solutions on platelets, Anesthesia and Analgesia 99 (2004), pp. 665–668. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (19)
72 T.J. Gan, E. Bennett-Guerrero and B. Phillips-Bute et al., Hextend, a physiologically balanced plasma expander for large volume use in major surgery: a randomized phase III clinical trial, Anesthesia and Analgesia 88 (1999), pp. 992–998. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (107)
73 J. Boldt, T. Schöllhorn and J. Münchbach et al., A total balanced volume replacement strategy using a new balanced hydroxyethyl starch preparation (6% HES 130/0.42) in patients undergoing major abdominal surgery, European Journal of Anaesthesiology 24 (2007), pp. 267–275. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
*74 M. Kulla, R. Weidhase and L. Lampl, Hydroxyethylstärke 6% 130/0,42 in Ringerazetat als Komponente eines balancierten Volumenersatzes in der Abdominalchirurgie, Anästhesiologie und Intensivmedizin 49 (2008), pp. 7–18. View Record in Scopus | Cited By in Scopus (1)
75 M. Wilkes, R. Navickis and W. Sibbald, Albumin versus hydroxyethyl starch in cardiopulmonary bypass surgery: a meta-analysis of postoperative bleeding, Annals of Thoracic Surgery 72 (2001), pp. 527–537.
*76 G. Haisch, J. Boldt and C. Krebs et al., The influence of intravascular volume therapy with a new hydroxyethyl starch preparation (6% HES 130/0.4) on coagulation in patients undergoing major abdominal surgery, Anesthesia and Analgesia 92 (2001), pp. 565–571. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (63)
77 J. Boldt, C. Knothe and B. Zickmann et al., Influence of different intravascular volume therapies on platelet function in patients undergoing cardiopulmonary bypass, Anesthesia and Analgesia 76 (1993), pp. 1185–1190. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (70)
78 S. Gandhi, R. Weiskopf and C. Jungheinrich et al., Volume replacement therapy during major orthopedic surgery using Voluven (hydroxyethyl starch 130/0.4) or hetastarch, Anesthesiology 106 (2007), pp. 1120–1127. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
*79 Kozek-Langenecker S, Jungheinrich C, Sauermann W, et al. Hydroxyethyl starch 130/0.4 (6%): effects on blood loss and use of blood products in major surgery – a pooled analysis of randomized clinical trials. Anesthesia and Analgesia 2008;107:382–390.
*80 D. Cheng, S. Belisle and M. Giffin et al., Colloids for perioperative plasma volume expansion: systematic review with meta-analysis of controlled trials, Transfusion Alternatives in Transfusion Medicine 9 (2007) 3 (abstract).
81 T. Haas, D. Fries and C. Holz et al., Less impairment of hemostasis and reduced blood loss in pigs after resuscitation from hemorrhagic shock using the small-volume concept with hypertonic saline/hydroxyethyl starch as compared to administration of 4% gelatin or 6% hydroxyethyl starch solution, Anesthesia and Analgesia 106 (2008), pp. 1078–1086. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
82 T. Standl, M. Burmeister and F. Schroeder et al., Hydroxyethyl starch (HES) 130/0.4 provides larger and faster increases in tissue oxygen tension in comparison with prehemodilution values than HES 70/0.5 or HES 200/0.5 in volunteers undergoing acute normovolemic hemodilution, Anesthesia and Analgesia 96 (2003), pp. 936–943. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (22)
83 N. Tabuchi, J. de Haan and R. Gallandat-Huet et al., Gelatin use impairs platelet adhesion during cardiac surgery, Thrombosis and Haemostasis 74 (1995), pp. 1447–1451. View Record in Scopus | Cited By in Scopus (43)
84 E. Mahla, T. Lang and M. Vincenzi et al., Thrombelastography for monitoring prolonged hypercoagulability after major abdominal surgery, Anesthesia and Analgesia 92 (2001), pp. 572–577. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)
*85 D.J. McCrath, E. Cerboni and R.J. Frumento et al., Thromboelastography maximum amplitude predicts postoperative thrombotic complications including myocardial infarction, Anesthesia and Analgesia 100 (2005), pp. 1576–1583. View Record in Scopus | Cited By in Scopus (45)
86 P. Collins, L. Macchiavello and S. Lewis et al., Global tests of haemostasis in critically ill patients with severe sepsis syndrome compared to controls, British Journal of Haematology 135 (2006), pp. 220–227. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (21)
87 L. Wang, J. Bastarache and L. Ware, The coagulation cascade in sepsis, Current Pharmaceutical Design 14 (2008), pp. 1860–1869. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
*88 H. Dieterich, T. Weissmüller and P. Rosenberger et al., Effect of hydroxyethyl starch on vascular leak syndrome and neutrophil accumulation during hypoxia, Critical Care Medicine 34 (2004), pp. 1775–1782.
89 K. Lang, S. Suttner and J. Boldt et al., Volume replacement with HES 130/0.4 may reduce the inflammatory response in patients undergoing major abdominal surgery, Canadian Journal of Anaesthesia 50 (2003), pp. 1009–1016. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (38)
90 A. Volta, V. Alvis and M. Campi et al., Influence of different strategies of volume replacement on the activity of matrix metalloproteinase: an in vitro and in vivo study, Anesthesiology 106 (2007), pp. 85–91.
91 P. Meyer, P. Pernet and B. Hejblum et al., Haemodilution induced by hydroxyethyl starches 130/0.4 is similar in septic and non-septic patients, Acta Anaesthesiologica Scandinavica 52 (2008), pp. 229–235. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
92 J.F. Hardy, P. de Moerloose and C.M. Samama, Massive transfusion and coagulopathy: pathophysiology and implications for clinical management, Canadian Journal of Anaesthesia 53 (2006), pp. S40–S58. View Record in Scopus | Cited By in Scopus (18)
93 S. Kozek-Langenecker, Management of massive operative blood loss, Minerva Anestesiologica 73 (2007), pp. 401–415. View Record in Scopus | Cited By in Scopus (19)
94 J. Watters, B. Tieu and J. Differding et al., A single bolus of 3% hypertonic saline with 6% dextran provides optimal initial resuscitation after uncontrolled hemorrhagic shock, The Journal of Trauma 61 (2006), pp. 75–81. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)
95 I. Wiedermann, G. Scharbert and H. Schöchl et al., Hydroxyethyl starch in hypertonic saline and platelet function, Anesthesiology 107 (2007), p. A549.
96 K. Brummel-Ziedins, M. Whelihan and E. Ziedins et al., The resuscitation fluid you choose may potentiate bleeding, The Journal of Trauma 61 (2006), pp. 1350–1358. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
97 N. Singh, S. Singh and L. Kaur et al., Morphological, thermal and rheological properties of starches from different botanical sources, Food Chemistry 81 (2003), pp. 219–231. Article | PDF (350 K) | View Record in Scopus | Cited By in Scopus (106)
98 I. Kwan, F. Bunn and F. Robert, Timing and volume of fluid administration for patients with bleeding, Cochrane Database of Systematic Reviews 3 (2003) CD002245.
99 P. Perel and I. Roberts, Colloids versus crystalloids for fluid resuscitation in critically ill patients, Cochrane Database of Systematic Reviews 4 (2008) CD000567.
100 S. Kozek-Langenecker and G. Scharbert, Which hemostatic changes determine clinical outcome?, Anesthesia and Analgesia 106 (2008), p. 1588. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
  相似文献   

6.
Volume kinetics could be a useful tool to better understand fluid distribution and its implications for fluid therapy. By using non-linear regression, it is possible to fit dilution data to an equation that describes fluid distribution in one- and two-volume models reasonably well. These models could be adapted to fit different clinical situations. In the future, it would be useful to design models similar to target control infusion models in pharmacokinetics. Such models should probably use dilution of Hb or other endogenous tracers in combination with blood pressure as endpoints.
• peri-operative fluid therapy continues to be an exercise in empiricism, with nagging questions about efficacy and complications
• in 1997, several authors introduced kinetic modeling (volume kinetics) describing the peak effects and clearance of intravenously infused fluids in terms similar to those used in pharmacokinetics
• this chapter briefly summarizes currently accepted principles of fluid therapy, discusses the general approach to kinetic analysis of fluid therapy, reviews currently available data defining kinetic responses to fluid therapy, and speculates about future applications of this approach
• further research is warranted where anaesthetic drugs in combination with haemorrhage are studied kinetically in humans
• it is important to implement a non-invasive analysis of an endogenous tracer such as Hb to facilitate future use of kinetic models for fluids
• an outcome study comparing fluid administration according to kinetic principles vs standard of care is warranted

References

1 G.T. Shires, J. Williams and F. Brown, Acute changes in extracellular fluid associated with major surgical procedures, Annals of Surgery 154 (1961), pp. 803–810.
*2 G.T. Shires, D. Coln, J. Carrico and S. Lightfoot, Fluid therapy in hemorrhagic shock, Archives of Surgery 88 (1964), pp. 688–693.
*3 B. Brandstrup, H. Tønnesen and R. Beier-Holgersen et al., Effects of intravenous fluid restriction on postoperative complications: comparison of two perioperative fluid regimens, Annals of Surgery 238 (2003), pp. 641–648. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (244)
4 L. Ståhle, A. Nilsson and R.G. Hahn, Modelling the volume of expandable body fluid spaces during i.v. fluid therapy, British Journal of Anaesthesia 78 (1997), pp. 138–143. View Record in Scopus | Cited By in Scopus (32)
*5 C. Svensen and R.G. Hahn, Volume kinetics of Ringer solution, dextran 70, and hypertonic saline in male volunteers, Anesthesiology 87 (1997), pp. 204–212. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (75)
6 R.G. Hahn and C. Svensen, Plasma dilution and the rate of infusion of Ringer's solution, British Journal of Anaesthesia 79 (1997), pp. 64–67. View Record in Scopus | Cited By in Scopus (26)
7 D.R. Stanski, The pharmacokinetics of intravenous fluids, Anesthesiology 87 (1997), pp. 200–201. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
8 F. Guarracino, F. Lapolla and C. Cariello et al., Target controlled infusion: TCI, Minerva Anestesiologica 71 (2005), pp. 335–337. View Record in Scopus | Cited By in Scopus (8)
9 M.C. van den Nieuwenhuyzen, F.H. Engbers, J. Vuyk and A.G. Burm, Target-controlled infusion systems: role in anaesthesia and analgesia, Clinical Pharmacokinetics 38 (2000), pp. 181–190. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
10 C.F. Minto, T.W. Schnider and K.M. Gregg et al., Using the time of maximum effect site concentration to combine pharmacokinetics and pharmacodynamics, Anesthesiology 99 (2003), pp. 324–333. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (39)
11 T.W. Schnider, A. Gentilini and R. Wymann, Feedback controlled anesthesia: can the computer replace the anesthesiologist?, Acta Anaesthesiologica Belgica 52 (2001), pp. 201–204. View Record in Scopus | Cited By in Scopus (1)
12 A. Gentilini, C.W. Frei and A.H. Glattfelder et al., Identification and targeting policies for computer-controlled infusion pumps, Critical Reviews in Biomedical Engineering 28 (2000), pp. 179–185. View Record in Scopus | Cited By in Scopus (8)
13 A.S. Tonnesen, Crystalloids and colloids. In: R.D. Miller, Editor, Anesthesia (4th edn.), Churchill Livingstone, New York, Edinburgh, London, Madrid, Melbounre, Milan, Tokyo (1994), pp. 1595–1617.
14 D.S. Prough and M. Mathru, Acid-base, fluids, and electrolytes. In: P.G. Barash, B.F. Cullen and R.K. Stoelting, Editors, Clinical anesthesia, Lippincott-Raven, Philadelphia (1997), pp. 157–187.
15 A.L. Cervera and G. Moss, Progressive hypovolemia leading to shock after continuous hemorrhage and 3:1 crystalloid replacement, American Journal of Surgery 129 (1975), pp. 670–674. Abstract | PDF (514 K) | View Record in Scopus | Cited By in Scopus (13)
16 L. Riddez, R.G. Hahn and B. Brismar et al., Central and regional hemodynamics during acute hypovolemia and volume substitution in volunteers, Critical Care Medicine 25 (1997), pp. 635–640. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (47)
17 B. Brandstrup, C. Svensen and A. Engquist, Hemorrhage and surgery cause a contraction of the extracellular space needing replacement – evidence and implications, Surgery 139 (2006), pp. 419–432. Article | PDF (202 K) | View Record in Scopus | Cited By in Scopus (17)
18 F.D. Moore, F.J. Dagher and C.M. Boyden et al., Hemorrhage in normal man: I. Distribution and dispersal of saline infusions following acute blood loss, Annals of Surgery 163 (1966), pp. 485–504. View Record in Scopus | Cited By in Scopus (21)
*19 R.G. Hahn, D.S. Prough and C. Svensen, Perioperative fluid management, Informa Healthcare USA, Inc., New York (2007).
20 S.B. Nadler, J.U. Hidalgo and T. Bloch, Prediction of blood volume in normal human adults, Surgery 51 (1962), pp. 224–232. View Record in Scopus | Cited By in Scopus (239)
21 Drobin D & Hahn RG. Time course of increased haemodilution in hypotension induced by extradural anaesthesia. British Journal of Anaesthesia 1996; 77: 223–226.
22 R.G. Hahn, Blood volume at the onset of hypotension in TURP performed during epidural anaesthesia, European Journal of Anaesthesiology 10 (1993), pp. 219–225. View Record in Scopus | Cited By in Scopus (8)
23 H. Chaplin, P.L. Mollison and H. Vetter, The body/venous hematocrit ratio: its constancy over a wide hematocrit range, The Journal of Clinical Investigation 32 (1953), pp. 1309–1316. Full Text via CrossRef
24 R.V. Ebert, E.A. Stead and J.G. Gibson, Response of normal subjects to acute blood loss, Archives of Internal Medicine 68 (1941), pp. 578–590.
25 H.J. Motulsky and L.A. Ransnas, Fitting curves to data using nonlinear regression: a practical and nonmathematical review, The FASEB Journal 1 (1987), pp. 365–374. View Record in Scopus | Cited By in Scopus (452)
26 D.S. Prough and J.D. Michenfelder, Cerebral blood flow and metabolism: implications for clinical monitoring, Critical care medicine: state of the art, Society of Critical Care Medicine, Fullerton, CA (1989), pp. 43–70.
27 H.G. Boxenbaum, S. Riegelman and R.M. Elashoff, Statistical estimations in pharmacokinetics, Journal of Pharmacokinetics and Biopharmaceutics 2 (1974), pp. 123–148. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (169)
*28 D. Drobin, A single-model solution for volume kinetic analysis of isotonic fluid infusions, Acta Anaesthesiologica Scandinavica 50 (2006), pp. 1074–1080. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
29 C. Svensen, K.I. Brauer and R. Hahn et al., Elimination rate constant describing clearance of 0.9% saline from plasma is independent of infused volume in sheep, Anesthesiology 101 (2004), pp. 666–674. View Record in Scopus | Cited By in Scopus (4)
30 Hahn RG. Increased haemodilution in hypotension induced by epidural anaesthesia. Acta Anaesthesiologica Scandinavica 1993; 37: 357–360.
31 R.G. Hahn, Origin of intravascular fluid recruited by vasodilatation during epidural anaesthesia, European Surgical Research 28 (1996), pp. 70–74. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (7)
32 K. Holte, N. Foss and C. Svensen et al., Epidural anesthesia and intravascular volume, Anesthesiology 100 (2004), pp. 281–286. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (31)
33 R.G. Hahn and M. Resby, Volume kinetics of Ringer's solution and dextran 3% during induction of spinal anaesthesia for caesarean section, Canadian Journal of Anaesthesia 45 (1998), pp. 443–451. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
34 Y. Li, S. Zhu and R.G. Hahn, The kinetics of Ringeŕs solution in young and elderly patients during induction of general anesthesia with propofol and epidural anesthesia with ropivacaine, Acta Anaesthesiologica Scandinavica 51 (2007), pp. 880–887. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
35 C.-A. Ewaldsson and R.G. Hahn, Volume kinetics if Ringer's solution during induction of spinal anaesthesia, British Journal of Anaesthesia 87 (2001), pp. 406–414. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (27)
36 C.-A. Ewaldsson and R.G. Hahn, Bolus injection of Ringeŕs solution and dextran 1 kDa during induction of spinal anesthesia, Acta Anaesthesiologica Scandinavica 49 (2005), pp. 152–159. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
*37 K.I. Brauer, C. Svensen and R.G. Hahn et al., Volume kinetic analysis of the distribution of 0.9% saline in conscious versus isoflurane-anesthetized sheep, Anesthesiology 96 (2002), pp. 442–449. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)
*38 C. Conolly, G.C. Kramer and R.G. Hahn et al., Isoflurane but not mechanical ventilation promotes third-space fluid losses during crystalloid volume loading, Anesthesiology 98 (2003), pp. 670–681.
*39 Å Norberg, K. Brauer and D. Prough et al., Volume turnover kinetics of fluid shifts in sheep after hemorrhage, fluid infusion and the combination of hemorrhage and fluid infusion, Anesthesiology 102 (2005), pp. 985–994. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (20)
40 C.-A. Ewaldsson and R.G. Hahn, Kinetics and extravascular retention of acetated Ringeŕs solution during isoflurane or propofol anesthesia for thyroid surgery, Anesthesiology 103 (2005), pp. 460–469. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (15)
41 R. Hahn, L.P. Brauer and P. Rodhe et al., Isoflurane inhibits transcapillary compensatory volume expansion, Anesthesia and Analgesia 103 (2006), pp. 350–358. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
42 C. Svensen, B. Clifton and K. Brauer et al., Sepsis produced by Pseudomonas bacteremia does not alter volume expansion after 0.9% saline infusion in sheep, Anesthesia and Analgesia 101 (2005), pp. 832–845.
43 M. Dubniks, J. Persson and P.-O. Grande, Effect of blood pressure on plasma volume loss in the rat under increased permeability, Intensive Care Medicine 33 (2007), pp. 2192–2198. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
44 L.A. Vane, D.S. Prough, C.A. Williams and G.C. Kramer, Effects of catecholamines on volume expansion and hemodynamic responses of crystalloid infusion, American Society of Anesthesiologists, Orlando, FL (2002) A678.
45 C. Svensen, S. Ponzer and R.G. Hahn, Volume kinetics of Ringer solution after surgery for hip fracture, Canadian Journal of Anaesthesia 46 (1999), pp. 133–141. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (18)
46 P. Strandberg and R.G. Hahn, Volume kinetics of glucose 2.5% solution and insulin resistance after abdominal hysterectomy, British Journal of Anaesthesia 94 (2005), pp. 30–38. View Record in Scopus | Cited By in Scopus (5)
*47 F. Sjöstrand and R.G. Hahn, Validation of volume kinetic analysis of glucose 2.5% solution given by intravenous infusion, British Journal of Anaesthesia 90 (2003), pp. 600–607. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (11)
*48 D. Drobin and R.G. Hahn, Volume kinetics of Ringer's solution in hypovolemic volunteers, Anesthesiology 90 (1999), pp. 81–91. Full Text via CrossRef
49 J. Olsson, C. Svensen and R.G. Hahn, The volume kinetics of acetated Ringer's solution during laparoscopic cholecystectomy, Anesthesia and Analgesia 99 (2004), pp. 1854–1860. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (10)
  相似文献   

7.
Walsh PC 《Urology》2001,57(6):1020-1024
I remain skeptical about the value of nerve grafts in restoring sexual function in men who undergo wide excision of the neurovascular bundle and am concerned that it may encourage urologists to pursue wide excision where it is neither necessary nor useful.
Patients who have invasion of the neurovascular bundles on both sides to the point where it is necessary to excise both neurovascular bundles are not curable with surgery.
Today, most men with localized prostate cancer who undergo surgery can have preservation of both neurovascular bundles.
Patients who have extensive disease outside the prostate are less likely to recover sexual function regardless of the status of their nerves and are unlikely to benefit from placement of a nerve graft.
A randomized properly performed study of nerve grafts should be carried out before nerve grafts are widely accepted in the management of localized prostate cancer.
The most valuable thing one can do to ensure the recovery of sexual function in a patient where it was necessary to widely excise one neurovascular bundle is not a nerve graft. Rather, it is precise preservation of the contralateral neurovascular bundle.

References

1. P.C. Walsh, Anatomic radical prostatectomy: evolution of the surgical technique. J Urol 160 (1998), pp. 2418–2424. Abstract | Article | PDF (1481 K) | Full Text via CrossRef
2. P.C. Walsh, Radical prostatectomy, preservation of sexual function, cancer control: the controversy. Urol Clin North Am 14 (1987), pp. 663–673. View Record in Scopus | Cited By in Scopus (59)
3. P.C. Walsh, J.I. Epstein and F.C. Lowe, Potency following radical prostatectomy with wide unilateral excision of the neurovascular bundle. J Urol 138 (1987), pp. 823–827. View Record in Scopus | Cited By in Scopus (70)
4. D.M. Quinlan, J.I. Epstein, B.S. Carter et al., Sexual function following radical prostatectomy: influence of preservation of neurovascular bundles. J Urol 145 (1991), pp. 998–1002. View Record in Scopus | Cited By in Scopus (247)
5. D.M. Quinlan, R.J. Nelson and P.C. Walsh, Cavernous nerve grafts to restore erectile function in a rat model. J Urol 141 (1989), p. 186A.
6. D.M. Quinlan, R.J. Nelson and P.C. Walsh, Cavernous nerve grafts restore erectile function in denervated rats. J Urol 145 (1991), pp. 380–383. View Record in Scopus | Cited By in Scopus (59)
7. J.K. Burgers, R.J. Nelson, D.M. Quinlan et al., Nerve growth factor, nerve grafts and amniotic membrane grafts restore erectile function in rats. J Urol 146 (1991), pp. 463–479.
8. D.M. Quinlan, R.J. Nelson, A.W. Partin et al., The rat as a model for the study of penile erection. J Urol 141 (1989), pp. 656–661. View Record in Scopus | Cited By in Scopus (74)
9. P.C. Walsh, Technique of vesicourethral anastomosis may influence recovery of sexual function following radical prostatectomy. Atlas Urol Clin North Am 2 (1994), pp. 59–63.
10. E.D. Kim, P.T. Scardino, O. Hampel et al., Interposition of sural nerve restores function of cavernous nerves resected during radical prostatectomy. J Urol 161 (1999), pp. 188–192. Abstract | Article | PDF (990 K) | Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (71)
11. J.I. Epstein, G. Pizov and P.C. Walsh, Correlation of pathologic findings with progression after radical retropubic prostatectomy. Cancer 71 (1993), pp. 3582–3593. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (436)
12. L. Cheng, M.F. Darson, E.J. Bergstralh et al., Correlation of margin status and extraprostatic extension with progression of prostate carcinoma. Cancer 86 (1999), pp. 1775–1782. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (105)
13. P.C. Walsh, Radical retropubic prostatectomy. In: P.C. Walsh, A.B. Retik, E.D. Vaughan, Jr et al.Campbell’s Textbook of Urology (7th ed.),, WB Saunders, Philadelphia (1998), pp. 2565–2588.
14. P.C. Walsh and P.J. Donker, Impotence following radical prostatectomy: insight into etiology and prevention. J Urol 128 (1982), pp. 492–497. View Record in Scopus | Cited By in Scopus (547)
15. H. Lepor, M. Gregerman, R. Crosby et al., Precise localization of the autonomic nerves from the pelvic plexus to the corpora cavernosa: a detailed anatomical study of the adult male pelvis. J Urol 133 (1985), pp. 207–212. View Record in Scopus | Cited By in Scopus (167)
16. M.A. Riopel, T.J. Polascik, A.W. Partin et al., Radical prostatectomy in men less than 50 years old. Urol Oncol 1 (1995), pp. 80–83. Abstract | Article | PDF (429 K) | View Record in Scopus | Cited By in Scopus (13)
17. G.F. Holmes, P.C. Walsh, C.R. Pound et al., Excision of the neurovascular bundle at radical prostatectomy in cases with perineural invasion on needle biopsy. Urology 53 (1999), pp. 752–756. Abstract | Article | PDF (451 K) | View Record in Scopus | Cited By in Scopus (34)
18. E.D. Kim, P.T. Scardino, D. Kadmon et al., Interposition sural nerve grafting during radical retropubic prostatectomy. Urology 57 (2001), pp. 211–216. Article | PDF (443 K) | View Record in Scopus | Cited By in Scopus (17)
19. C.R. Pound, A.W. Partin, J.I. Epstein et al., Prostate-specific antigen after anatomic radical retropubic prostatectomy. Urol Clin North Am 24 (1997), pp. 395–406. Abstract | Article | PDF (756 K) | View Record in Scopus | Cited By in Scopus (377)
20. F. Rabbani, A.M. Stapleton, T.M. Wheeler et al., Predicting recovery of erectile potency after radical prostatectomy: influence of age, preoperative potency, and extent of nerve-sparing. J Urol 163 suppl (2000), p. 287.
  相似文献   

8.
9.
During cardiac surgery procedures, the first item to consider is whether the surgery will be performed with or without CPB, because its use implies the utilization of heat exchange and the possibility of controlling the core body temperature.Although irrefutable advantages of the use of hypothermia instead of normothermia during CPB have not been reported, it seems wise to maintain a mild core body temperature (34–35 °C) in order to preserve brain function during potential ischaemic periods due to intra-operative incidences. Beyond the objective of maintaining a specific core temperature, it is much more important, from the point of view of overall postoperative brain outcome, to avoid brain hyperthermia during the rewarming period at the end of CPB. This hyperthermia is secondary to fast rewarming, with uncontrolled arterial temperature infusion ≥39 °C trying to reduce the development of post-CPB hypothermia as a consequence of afterdrop. Brain hyperthermia is highly prevalent in cardiac surgery, but is often poorly understood and not taken into account. The main reason for this is the absence of specific monitoring to detect brain desaturation and ischaemia.The authors propose the use of a mild temperature during CPB, combined with active heat transfer by application of a peripheral on the body surface. This reduces the need for intensive rewarming with closer values for the core and peripheral temperatures.

Epilogue

As reported by Flynn et al63, available reports are flawed by inconsistencies in cardioplegic technique and inadequate definitions of normothermic systemic perfusion. These flaws make definition of the independent effects of warm blood cardioplegia and normothermic CPB uncertain. The American College of Cardiology/American Heart Association (ACC/AHA) guidelines for CABG (1999) reported that ‘no strong argument can currently be made for warm versus cold and crystalloid versus blood cardioplegia’ in patients with normal left ventricular function. The guidelines go on to report ‘certain techniques may offer a wider margin of safety for special patient subsets’. However, the evidence for such a wide safety margin is documented only as: ‘Several studies have suggested that blood cardioplegia (compared to crystalloid) may offer a greater margin of safety during CABG performed on patients with acute coronary occlusion, failed angioplasty, urgent re-vascularisation for unstable angina, and/or chronically impaired left ventricular function’. According to the 2005 update, ‘additional manoeuvres to reduce type 2 neurological injury include maintenance of steady cerebral blood flow during CPB, avoidance of cerebral hyperthermia during and after CPB, meticulous control of perioperative hyperglycemia, and avoidance and limitation of post-operative cerebral oedema’.73Finally, it should be remembered that the cost of preventing intra-operative hypothermia is much less than the cost of treating its adverse consequences.74 Moreover, in cases where some type of CPB is performed, warming of the peripheral compartment reduces the afterdrop and the need for intensive rewarming, thus reducing the probability of inducing brain hyperthermia, the cause of many deleterious effects.33
• Nowadays there is no consistent clear advantage of cold over warm of normothermic CPB.
• Mild brain cooling (2–5 °C) confers protection from ischaemic brain injury, and reduces infarct size in transient middle artery occlusion. So it seems wise to maintain a mild core body temperature (34–35 °C) during CPB.
• Maintenance of normothermia throughout the non-CPB phases of CABG is associated with reduced myocardial ischaemic injury, lower SVR and higher cardiac index.
• If core temperature is preserved but skin temperature is lowered, local bleeding time increases.
• The strict avoidance of hyperthermia during rewarming period is mandatory, because it could exacerbate brain injury.
• Mild temperature during CPB, combined with warming of the peripheral compartment reduces the afterdrop and the need for intensive rewarming, thus reducing the probability of inducing brain hyperthermia in addition to postoperative hypothermia.
• Till now, we do not know which is the most appropriate temperature for CPB during cardiac surgery, in terms of improving brain and no brain overall perioperative outcome.
• Further studies are needed to find a more accurate way of monitoring brain temperature and the possible relationship between brain hyperthermia and postoperative neurocognitive deficits.
• More efforts are required to determine the effects of hypothermia on drug disposition and response, and thus delineate better dosing guidelines.
• Further studies are needed to clarify which is the more efficient warm system to maintain the patients actively warmed during OPCAB.

References

1 DeFrances CJ & Hall MJ. National Hospital Discharge Survey. CDC Advance Data 2007. Atlanta, USA: U.S. Department of Health and Human Services, 2005. Available from: http://www.cdc.gov/nchs/data/ad/ad385.pdf.
*2 D.J. Cook, Changing temperature management for cardiopulmonary bypass, Anesth Analg 88 (1999), pp. 1254–1271. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (39)
3 A.R. Metha, M.E. Romanoff and M.G. Licina, Anesthetic management in the precardiopulmonary bypass period. In: F.A. Hensley, D.E. Martin and G.P. Gravlee, Editors, A practical approach to Cardiac Anesthesia (4th edn.), Lippincott Willians & Wilkins, Philadelphia (2008), pp. 176–197.
4 G.J. Pestel and A. Kurz, Hypothermia – it's more than a toy, Curr Opin Anesthesiol 18 (2005), pp. 151–156. View Record in Scopus | Cited By in Scopus (7)
5 C.D. Deakinn, F. Clewlow and T. Pierce, Thermal energy balance as a measure of adequate rewarming from hypothermic cardiopulmonary bypass, Journal of Cardiothoracic and Vascular Anesthesia 14 (2000), pp. 388–392.
6 A. Kurz, D.I. Sessler and R.A. Lenhardt, Study of Wound Infections and Temperature Group. Perioperative normothermia to reduce the incidence of surgical-wound infection and shorten hospitalization, NEJM 334 (1996), pp. 1209–1215. View Record in Scopus | Cited By in Scopus (807)
7 J. Boldt, C. Knothe and B. Zickmann et al., Platelet function in cardiac surgery: influence of temperature and aprotinin, Ann Thorac Surg 55 (1993), pp. 652–658. View Record in Scopus | Cited By in Scopus (54)
8 R.M. Engelman, B. Pleet and J.A. Rousou et al., Influence of cardiopulmonary bypass perfusion temperature on neurologic and hematologic function after coronary artery bypass grafting, Ann Thorac Surg 67 (1999), pp. 1547–1556. View Record in Scopus | Cited By in Scopus (36)
9 P.M. Honore, L.M. Jacquet and R.J. Beale et al., Effects of normothermia versus hypothermia on extravascular lung water and serum cytokines during cardiopulmonary bypass: a randomized, controlled trial, Crit Care Med 29 (2001), pp. 1903–1909. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (28)
10 P. Khatri, P. Babyak and N.D. Croughwell et al., Temperature during coronary artery bypass surgery affects quality of life, Ann Thorac Surg 71 (2001), pp. 110–116. Abstract | Article | PDF (216 K) | View Record in Scopus | Cited By in Scopus (9)
11 G.T. Christakis, J.P. Koch and K.A. Deemar et al., A randomized study of the systemic effects of warm heart surgery, Ann Thorac Surg 54 (1992), pp. 449–459. View Record in Scopus | Cited By in Scopus (71)
12 W.M. Brown, J.L. Jay and J.P. Gott et al., Warm blood cardioplegia: superior protection after acute myocardial ischemia, Ann Thorac Surg 55 (1993), pp. 32–41.
13 Y.J. Woo, P. Atluri and T.J. Grand et al., Active thermoregulation improves outcome of off-pump coronary artery bypass, Asian Cardiovasc Thorac Ann 13 (2005), pp. 157–160. View Record in Scopus | Cited By in Scopus (6)
14 B. Romlin, K. Petruson and K. Nilsson, Moderate superficial hypothermia prolongs bleeding time in humans, Acta Anaesthesiol Scand 51 (2007), pp. 198–201. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
15 I. Birdi, C.M. Caputo and G.D. Angelini et al., Influence of normothermic systemic perfusion temperature on cold myocardial protection during coronary artery bypass, Cardiovascular Surgery (London, England) 7 (1999), pp. 369–374. Abstract | View Record in Scopus | Cited By in Scopus (10)
*16 R.M. Engelman, A.B. Pleet and R. Hicks et al., Is there a relationship between systemic perfusion temperature during coronary artery bypass grafting and extent of intraoperative ischemic central nervous system injury?, The Journal of Thoracic and Cardiovascular Surgery 119 (2000), pp. 230–232. Abstract | Article | PDF (21 K) | View Record in Scopus | Cited By in Scopus (13)
17 A. Lavinio, I. Timofeev and J. Nortje et al., Cerebrovascular reactivity during hypothermia and rewarming, Br J Anaesth 99 (2007), pp. 237–244. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)
18 S.R. Insler, M.S. O'Connor and M.J. Leventhal et al., Association between postoperative hypothermia and adverse outcome after coronary artery bypass surgery, Ann Thorac Surg 70 (2000), pp. 175–181. Abstract | Article | PDF (310 K) | View Record in Scopus | Cited By in Scopus (33)
*19 H.J. Nathan, L. Parlea and J.Y. Dupuis et al., Safety of deliberate intraoperative and postoperative hypothermia for patients undergoing coronary artery surgery: a randomized trial, J Thorac Cardiovasc Surg 127 (2004), pp. 1270–1275. Abstract | Article | PDF (91 K) | View Record in Scopus | Cited By in Scopus (25)
20 T. Heier and J.E. Caldwell, Impact of hypothermia on the response to neuromuscular blocking drugs, Anesthesiology 104 (2006), pp. 1070–1080. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (9)
21 G.W. Roach, M. Kanchuguer and C.M. Mangano et al., Adverse cerebral outcomes after coronary bypass surgery, N Eng J Med 335 (1996), pp. 1857–1863. View Record in Scopus | Cited By in Scopus (938)
22 M.S. Ali, M. Harmer and F. Kirkham, Cardiopulmonary bypass temperature and brain function, Anaesthesia 60 (2005), pp. 365–372.
23 Y. Kadoi, F. Kawahara and S. Saito et al., Effects of hypothermic and normothermic cardiopulmonary bypass on brain oxygenation, Ann Thorac Surg 68 (1999), pp. 34–39. Abstract | Article | PDF (219 K) | View Record in Scopus | Cited By in Scopus (32)
24 K.G. Shann, D.S. Likosky and J.M. Murkin et al., An evidence-based review of the practice of cardiopulmonary bypass in adults: a focus on neurologic injury, glycemic control, hemodilution, and the inflammatory response, J Thorac Cardiovasc Surg 132 (2006), pp. 283–290.
25 R.F. MacLean, B.I. Wong and C.D. Naylor et al., Cardiopulmonary bypass temperature and central nervous system dysfunction, Circulation 90 (1994), pp. II250–II255.
26 H.J. Nathan, G.A. Wells and J.L. Munson et al., Neuroprotective effect of mild hypothermia in patients undergoing coronary artery surgery with cardiopulmonary bypass, Circulation 104 (2001), pp. I85–I91.
27 M. Boodhwani, F.D. Rubens and D. Wozny et al., Predictors of early neurocognitive deficits in low-risk patients undergoing on-pump coronary artery bypass surgery, Circulation 114 (Suppl. 1) (2006), pp. I461–I466. View Record in Scopus | Cited By in Scopus (10)
28 M. Boodhwani, F. Rubens and D. Denise Wozny et al., Effects of sustained mild hypothermia on neurocognitive function after coronary artery bypass surgery: a randomized, double-blind study, J Thorac Cardiovasc Surg 134 (2007), pp. 1443–1452.
29 G. DeFoe, C. Krumholz and C. DioDato et al., Lowest core body temperature and adverse outcomes associated with coronary artery bypass surgery, Perfusion 18 (2003), pp. 127–133. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (1)
30 K. Rees, M. Beranek-Stanley and M. Burke et al., Hypothermia to reduce neurological damage following coronary artery bypass surgery, Cochrane Database System Reviews 1 (2001) CD002138.
*31 Y. Kadoi and F. Goto, Factors associated with postoperative cognitive dysfunction in patients undergoing cardiac surgery, Surg Today 36 (2006), pp. 1053–1057. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3)
32 P.M. Ho, D.B. Arciniegas and J. Grigsby et al., Predictors of cognitive decline following coronary artery bypass graft surgery, Ann Thorac Surg 77 (2004), pp. 597–603. Abstract | Article | PDF (94 K) | View Record in Scopus | Cited By in Scopus (25)
33 G. Dupuis, E. Kennedy and R. Lindquist et al., Coronary artery bypass graft surgery and cognitive performance, Am J Crit Care 15 (2006), pp. 471–479. View Record in Scopus | Cited By in Scopus (3)
34 A.M. Grigore, H.P. Grocott and J.P. Matthew et al., The rewarming rate and increased peak temperature alter neurocognitive outcome after cardiac surgery, Anesth Analg 94 (2002), pp. 4–10. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (69)
*35 H. Kaukuntla, D. Harrington and I. Bilkoo et al., Temperature monitoring during cardiopulmonary bypass – do we undercool or overheat the brain?, Eur J Cardiothorac Surg 26 (2004), pp. 580–585. Article | PDF (191 K) | View Record in Scopus | Cited By in Scopus (15)
36 F. Kawhara, Y. Kadoi and F. Goto et al., Slow rewarming improves jugular venous oxygen saturation during rewarming, Acta Anaesthesiol Scand 47 (2003), pp. 419–424.
*37 N. Nesher, Eli Zisman and T. Wolf et al., Strict thermoregulation attenuated myocardial injury during coronary artery bypass graft surgery as reflected by reduced levels of cardiac-specific troponin I, Anesth Analg 96 (2003), pp. 328–335. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
38 D. Myron, M. Ginsberg and R. Busto, Combating hyperthermia in acute stroke: a significant clinical concern, Stroke 29 (1998), pp. 529–534.
39 N.A. Nussmeier, W. Cheng and M. Marino et al., Temperature during cardiopulmonary bypass: the discrepancies between monitored sites, Anesth Analg 103 (2006), pp. 1373–1379. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
*40 S. Bar-Yosef, J.P. Mathew and M.F. Newman et al., Prevention of cerebral hyperthermia during cardiac surgery by limiting on-bypass rewarming in combination with post-bypass body surface warming: a feasibility study, Anesth Analg 99 (2004), pp. 641–646. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
41 F. Puskas, H.P. Grocott and W.D. White et al., Intraoperative hyperglycemia and cognitive decline after CABG, Ann Thorac Surg 84 (2007), pp. 1467–1473. Abstract | Article | PDF (134 K) | View Record in Scopus | Cited By in Scopus (15)
42 C. Naughton, N. Reilly and A. Powroznyk, Factors determining the duration of tracheal intubation in cardiac surgery: a single-centre sequential patient audit, Eur J Anaesthesiol 20 (2003), pp. 225–233. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (8)
43 D.I. Sessler, Heat preservation during cardiac surgery, J Cardiothorac Vasc Anesth 14 (2000), pp. 499–500. Abstract | PDF (21 K) | View Record in Scopus | Cited By in Scopus (1)
44 D.I. Sessler, Hyperthermia after cardiac surgery: mechanism and importance, J Cardiothorac Vasc Anesth 19 (2005), pp. 423–425. Article | PDF (70 K) | View Record in Scopus | Cited By in Scopus (3)
45 H.P. Grocott, G.B. Mackensen and A.M. Grigore et al., Postoperative hyperthermia is associated with cognitive dysfunction after coronary artery bypass graft surgery, Stroke 33 (2002), pp. 537–541. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (62)
46 M. Licker, Schaweizer and F.E. Ralley, Thermoregulatory and metabolic responses following cardiac surgery, Eur J Anaesthesiol 13 (1996), pp. 502–510. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
47 A. Parolari, F. Alamanni and G. Polvani et al., Meta-analysis of randomized trials comparing off-pump with on-pump coronary artery bypass, Ann Thorac Surg 80 (2005), pp. 2121–2125. Abstract | Article | PDF (108 K) | View Record in Scopus | Cited By in Scopus (46)
48 D.C. Cheng, D. Brainbridge and J.E. Martin et al., Does off-pump coronary artery bypass reduce mortality, morbidity, and resource utilization when compared with conventional coronary artery bypass? A meta-analysis of randomized trials, Anesthesiology 102 (2005), pp. 188–203. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (159)
*49 G. van der Heijden, H. Nathoe and E. Jansen et al., Meta-analysis on the effect of off-pump coronary bypass surgery, Eur J Cardiothorac Surg 26 (2004), pp. 81–84. Article | PDF (95 K) | View Record in Scopus | Cited By in Scopus (41)
50 A.C. St. André and A. DelRossi, Hemodynamic management of patients in the first 24 hours after cardiac surgery, Crit Care Med 33 (2005), pp. 2082–2093. View Record in Scopus | Cited By in Scopus (14)
51 T. Stanley, H. Grocott and B. Phillips-Bute et al., Preliminary evaluation of the Arctic Sun temperature-controlling system during off-pump coronary artery bypass surgery, Ann Thorac Surg 75 (2003), pp. 1140–1144. Abstract | Article | PDF (139 K) | View Record in Scopus | Cited By in Scopus (11)
52 T. Vassiliades, J. Nielsen and J. Lonqquist, Evaluation of a new temperature management system during off-pump coronary artery bypass, Interact CardioVasc Thorac Surg 2 (2003), pp. 454–457. Abstract | View Record in Scopus | Cited By in Scopus (3)
53 N. Nesher, T. Wolf and I. Kushnir et al., Novel thermoregulation system for enhancing cardiac function and hemodynamics during coronary artery bypass graft surgery, Ann Thorac Surg 72 (2001), pp. S1069–S1076. Abstract | Article | PDF (1229 K) | View Record in Scopus | Cited By in Scopus (13)
54 A.H. Kiessling, F. Isgro and A. Lehmann et al., Evaluating a new method for maintaining body temperature during OPCAB and robotic procedures, Med Sci Monit 12 (2006), pp. 39–42.
55 C.K. Hofer, M. Worn and R. Tavaloki et al., Influence of body core temperature on blood loss and transfusion requirements during off-pump coronary artery bypass grafting: a comparison of 3 warming systems, J Thorac Cardiovasc Surg 129 (2005), pp. 838–843. Abstract | Article | PDF (204 K) | View Record in Scopus | Cited By in Scopus (17)
56 A. Bräuer, M.J. English and N. Steinmetz et al., Efficacy of forced-air warming systems with full body blankets, Can J Anesth 54 (2007), pp. 34–41. View Record in Scopus | Cited By in Scopus (6)
57 A. Wadhwa, R. Komatsu and M. Orhan-Sungur et al., New circulating-water devices warm more quickly than forced-air in volunteers, Anesth Analg 105 (2007), pp. 1681–1687. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (5)
58 A. Rajek, R. Lenhardt and D.I. Sessler et al., Efficacy of two methods for reducing postbypass afterdrop, Anesthesiology 92 (2000), pp. 447–456. Full Text via CrossRef
59 J.Y. Kim, H. Shinn and Y.J. Oh et al., The effect of skin surface warming during anesthesia preparation on preventing redistribution hypothermia in the early operative period of off-pump coronary artery bypass surgery, Eur J Cardiothorac Surg 29 (2006), pp. 343–347. Article | PDF (94 K) | View Record in Scopus | Cited By in Scopus (10)
*60 F.F. Immer, A. Ackermannn and E. Cygax et al., Minimal extracorporeal circulation is a promising technique for coronary artery bypass grafting, Ann Thorac Surg 84 (2007), pp. 1515–1521. Abstract | Article | PDF (183 K) | View Record in Scopus | Cited By in Scopus (16)
61 D.H. Adams, E.J. Heyer and A.E. Simon et al., Incidence of atrial fibrillation after mild or moderate hypothermic cardiopulmonary bypass, Crit Care Med 28 (2000), pp. 309–311. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (23)
62 L.L. Creswell, J.C. Alexander and T.B. Ferguson et al., Intraoperative interventions. American College of Chest Physicians Guidelines for the prevention and management of postoperative atrial fibrillation after cardiac surgery, Chest 128 (2005), pp. 28S–35S. Full Text via CrossRef
63 K. Flynn, E. Adams and E. Alligood, Optimal Temperature for Cardioplegia during Coronary Artery Bypass Grafting. Department of Veterans Affairs, 2003 Available from: http://www.va.gov/vatap/pubs/cardioplegia.pdf (2003).
64 A. Kapetanopoulos, M.C. Katsetos and J. Kluger, Intraoperative hypothermia increased defibrillation energy requirements, The Journal of Cardiovascular Medicine 8 (2007), pp. 741–743. View Record in Scopus | Cited By in Scopus (1)
65 T.O. Stanley, G.B. Mackensen and H.P. Grocott et al., The impact of postoperative atrial fibrillation on neurocognitive outcome after coronary artery bypass graft surgery, Anesth Analg 94 (2002), pp. 290–295. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)
*66 A. Gega, J.A. Rizzo and M.H. Johnson et al., Straight deep hypothermic arrest: experience in 394 patients support its effectiveness as a sole mean of brain preservation, Ann Thorac Surg 84 (2007), pp. 759–767. Abstract | Article | PDF (748 K) | View Record in Scopus | Cited By in Scopus (16)
67 R.B. Griepp, Cerebral protection during aortic arch surgery, J Thorac Cardiovasc Surg 121 (2001), pp. 425–427. Abstract | PDF (45 K) | View Record in Scopus | Cited By in Scopus (52)
68 A. Rajek, R. Lenhardt and D.I. Sessler et al., Tissue heat content and distribution during and after cardiopulmonary bypass at 31 °C and 27 °C, Anesthesiology 88 (1998), pp. 1511–1518. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (36)
*69 A. Rajek, R. Lenhardt and D.I. Sessler et al., Tissue heat distribution during and after cardiopulmonary bypass at 17 °C, Anesth Analg 88 (1999), pp. 1220–1225. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (13)
70 R.A. Peragallo and D.C.H. Cheng, Tracheal extubation should not occur routinely in the operating room after cardiac surgery, J Cardiothorac Vasc Anesth 14 (2000), pp. 611–613. Abstract | PDF (33 K) | View Record in Scopus | Cited By in Scopus (7)
71 P.E. Stensrud, G.A. Nuttall and M. de Castro et al., A prospective, randomized study of cardiopulmonary bypass temperature and blood transfusion, Ann Thorac Surg 66 (1999), pp. 711–715. Abstract | Article | PDF (189 K) | View Record in Scopus | Cited By in Scopus (19)
72 M.A. Tortorici, P.M. Kochanek and S.M. Poloyac, Effects of hypothermia on drug disposition, metabolism, and response: a focus of hypothermia-mediated alterations on the cytochrome P450 enzyme system, Crit Care Med 35 (2007), pp. 2196–2204. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (26)
73 , ACC/AHA Pocket Guideline Coronary Artery Bypass Graft Surgery, American College of Cardiology Foundation and American Heart Association, Inc, Washington, DC (2005) Available from: http://www.acc.org/qualityandscience/clinical/guidelines/cabg/index_pkt.pdf.
74 E.M. Scott and R. Buckland, A systematic review of intraoperative warming to prevent postoperative complications, AORN J 83 (2006), pp. 1090–1113. Abstract | Article | PDF (551 K) | View Record in Scopus | Cited By in Scopus (7)
  相似文献   

10.
The incidence of DSWI after cardiac surgery is approximately 1.1% with an associated mortality of 13%. These numbers have changed little in the last 20 years despite numerous advances in the care of patients undergoing cardiac surgery. This is likely a reflection of an older patient population with more co-morbidities in the modern era. Many of the risk factors for DSWI are not modifiable. However, treating these diseases when possible, particularly glucose intolerance with insulin infusions seems beneficial in reducing the incidence of DSWI. There is active investigation into the utility of eradicating S. aureus from the naso-pharyngeal and skin flora. To this end, mupirocin and chlorhexidine gluconate may prove beneficial. While there has been some interest in using glycopeptides (vancomycin) for surgical prophylaxis in areas where MRSA infections are highly prevalent, this practice is not supported by any evidence based literature.
• The use of insulin infusions to maintain glucose levels < 200 mg/dl perioperatively appears to be beneficial in preventing DSWIs. However, very tight intraoperative glucose control (80–100 mg/dl) may increase the risk of stroke and death.
• Eradication of Staphylococcus aureus from the naso-pharygeal flora with topical mupirocin and/ or chlorhexidine oral rinse is a low risk intervention that may decrease the incidence of DSWI as well as antibiotic usage
• It appears that even in hospitals with a high incidence of methicillin resistant S. aureus surgical site infections first generation cephalosporins (cefazolin) are superior to glycopeptides (vancomycin) for surgical prophylaxis.
• The low incidence of DSWIs makes the performance of appropriately powered, randomized trials difficult.
• Large, multi-center trials are needed to evaluate the efficacy of interventions such as the eradication of S. aureus from the nasopharynx in preventing DSWIs.
• Like many complications from cardiac surgery including neurocognitive dysfunction and renal impairment, there is likely no “silver bullet” that will prevent DSWIs. As such, the focus should be on system-based approaches that incorporate multiple modalities of prevention.

References

1 C.Y. Bitkover and B. Gardlund, Mediastinitis after cardiovascular operations: a case-control study of risk factors, Annals of Thoracic Surgery 65 (1998), pp. 36–40. Abstract | Article | PDF (225 K) | View Record in Scopus | Cited By in Scopus (79)
2 M. De Feo, A. Renzulli and G. Ismeno et al., Variables predicting adverse outcome in patients with deep sternal wound infection, Annals of Thoracic Surgery 71 (2001), pp. 324–331. Abstract | Article | PDF (173 K) | View Record in Scopus | Cited By in Scopus (42)
3 R.M. El Oakley and J.E. Wright, Postoperative mediastinitis: classification and management, Annals of Thoracic Surgery 61 (1996), pp. 1030–1036. Abstract | PDF (773 K) | View Record in Scopus | Cited By in Scopus (193)
4 D. Jonkers, T. Elenbaas and P. Terporten et al., Prevalence of 90-days postoperative wound infections after cardiac surgery, European Journal of Cardio-Thoracic Surgery 23 (2003), pp. 97–102. Article | PDF (131 K) | View Record in Scopus | Cited By in Scopus (32)
5 P. Munoz, A. Menasalvas and J.C. Bernaldo de Quiros et al., Postsurgical mediastinitis: a case-control study, Clinical Infectious Diseases 25 (1997), pp. 1060–1064. View Record in Scopus | Cited By in Scopus (48)
6 A. Salehi Omran, A. Karimi and S.H. Ahmadi et al., Superficial and deep sternal wound infection after more than 9000 coronary artery bypass graft (CABG): incidence, risk factors and mortality, BMC Infectious Disease 7 (2007), p. 112. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (6)
7 M. Sharma, D. Berriel-Cass and J. Baran Jr., Sternal surgical-site infection following coronary artery bypass graft: prevalence, microbiology, and complications during a 42-month period, Infection Control and Hospital Epidemiology 25 (2004), pp. 468–471. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (24)
8 R. Wouters, F. Wellens and H. Vanermen et al., Sternitis and mediastinitis after coronary artery bypass grafting. Analysis of risk factors, Texas Heart Institute Journal 21 (1994), pp. 183–188. View Record in Scopus | Cited By in Scopus (30)
*9 W.J. Mauermann and E.C. Nemergut, The anesthesiologist's role in the prevention of surgical site infections, Anesthesiology 105 (2006), pp. 413–421 quiz 39–40. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (12)
10 W. Rosamond, K. Flegal and K. Furie et al., Heart disease and stroke statistics–2008 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee, Circulation 117 (2008), pp. e25–e146.
11 I.K. Toumpoulis, C.E. Anagnostopoulos and J.J. Derose Jr. et al., The impact of deep sternal wound infection on long-term survival after coronary artery bypass grafting, Chest 127 (2005), pp. 464–471. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (46)
12 , Risk factors for deep sternal wound infection after sternotomy: a prospective, multicenter study, The Journal of Thoracic and Cardiovascular Surgery 111 (1996), pp. 1200–1207.
13 P. Centofanti, F. Savia and M. La Torre et al., A prospective study of prevalence of 60-days postoperative wound infections after cardiac surgery. An updated risk factor analysis, The Journal of Cardiovascular Surgery 48 (2007), pp. 641–646. View Record in Scopus | Cited By in Scopus (1)
*14 , Guidelines and Indications For Coronary Artery Bypass Surgery. A Report Of The American College of Cardiolgy/ American Heart AssociationTask Force on Assessment of Diagnositc and Therapeutic Cardiovascular Procedures, Journal of the American College of Surgeons 17 (1991), pp. 543–589.
15 F.D. Loop, B.W. Lytle and D.M. Cosgrove et al., Influence of the internal-mammary-artery graft on 10-year survival and other cardiac events, The New England Journal of Medicine 314 (1986), pp. 1–6. View Record in Scopus | Cited By in Scopus (995)
16 M. Arnold, The surgical anatomy of sternal blood supply, The Journal of Thoracic and Cardiovascular Surgery 64 (1972), pp. 596–610. View Record in Scopus | Cited By in Scopus (54)
17 A.E. Seyfer, C.D. Shriver and T.R. Miller et al., Sternal blood flow after median sternotomy and mobilization of the internal mammary arteries, Surgery 104 (1988), pp. 899–904. View Record in Scopus | Cited By in Scopus (65)
18 E.A. Grossi, R. Esposito and L.J. Harris et al., Sternal wound infections and use of internal mammary artery grafts, The Journal of Thoracic and Cardiovascular Surgery 102 (1991), pp. 342–346 discussion 6–7.
19 S.R. Hazelrigg, H.A. Wellons Jr. and J.A. Schneider et al., Wound complications after median sternotomy. Relationship to internal mammary grafting, The Journal of Thoracic and Cardiovascular Surgery 98 (1989), pp. 1096–1099. View Record in Scopus | Cited By in Scopus (52)
20 N.T. Kouchoukos, T.H. Wareing and S.F. Murphy et al., Risks of bilateral internal mammary artery bypass grafting, Annals of Thoracic Surgery 49 (1990), pp. 210–217 discussion 7–9.
21 D.L. Galbut, E.A. Traad and M.J. Dorman et al., Seventeen-year experience with bilateral internal mammary artery grafts, Annals of Thoracic Surgery 49 (1990), pp. 195–201. View Record in Scopus | Cited By in Scopus (126)
22 G.W. He, W.H. Ryan and T.E. Acuff et al., Risk factors for operative mortality and sternal wound infection in bilateral internal mammary artery grafting, The Journal of Thoracic and Cardiovascular Surgery 107 (1994), pp. 196–202. View Record in Scopus | Cited By in Scopus (66)
23 F.D. Loop, B.W. Lytle and D.M. Cosgrove et al., J. Maxwell Chamberlain memorial paper. Sternal wound complications after isolated coronary artery bypass grafting: early and late mortality, morbidity, and cost of care, Annals of Thoracic Surgery 49 (1990), pp. 179–186 discussion 86–7.
*24 F. Filsoufi, P.B. Rahmanian and J.G. Castillo et al., Incidence, treatment strategies and outcome of deep sternal wound infection after orthotopic heart transplantation, The Journal of Heart and Lung Transplantation 26 (2007), pp. 1084–1090. Abstract | Article | PDF (167 K) | View Record in Scopus | Cited By in Scopus (2)
25 M. Carrier, L.P. Perrault and M. Pellerin et al., Sternal wound infection after heart transplantation: incidence and results with aggressive surgical treatment, Annals of Thoracic Surgery 72 (2001), pp. 719–723 discussion 23–4. Abstract | Article | PDF (327 K) | View Record in Scopus | Cited By in Scopus (17)
26 M. Senechal, P. LePrince and S. Tezenas du Montcel et al., Bacterial mediastinitis after heart transplantation: clinical presentation, risk factors and treatment, The Journal of Heart and Lung Transplantation 23 (2004), pp. 165–170. Abstract | Article | PDF (75 K) | View Record in Scopus | Cited By in Scopus (12)
27 R.H. Breyer, S.A. Mills and A.S. Hudspeth et al., A prospective study of sternal wound complications, Annals of Thoracic Surgery 37 (1984), pp. 412–416. View Record in Scopus | Cited By in Scopus (44)
28 T.L. Demmy, S.B. Park and G.A. Liebler et al., Recent experience with major sternal wound complications, Annals of Thoracic Surgery 49 (1990), pp. 458–462. View Record in Scopus | Cited By in Scopus (87)
29 L.S. Newman, L.C. Szczukowski and R.P. Bain et al., Suppurative mediastinitis after open heart surgery. A case control study of risk factors, Chest 94 (1988), pp. 546–553. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (47)
30 J.J. Curtis, N.C. Clark and C.A. McKenney et al., Tracheostomy: a risk factor for mediastinitis after cardiac operation, Annals of Thoracic Surgery 72 (2001), pp. 731–734. Abstract | Article | PDF (165 K) | View Record in Scopus | Cited By in Scopus (22)
31 S.D. Force, D.L. Miller and R. Petersen et al., Incidence of deep sternal wound infections after tracheostomy in cardiac surgery patients, Annals of Thoracic Surgery 80 (2005), pp. 618–621 discussion 21–2.
*32 P.B. Rahmanian, D.H. Adams and J.G. Castillo et al., Tracheostomy is not a risk factor for deep sternal wound infection after cardiac surgery, Annals of Thoracic Surgery 84 (2007), pp. 1984–1991. Abstract | Article | PDF (105 K) | View Record in Scopus | Cited By in Scopus (8)
33 J.L. Diehl, S. El Atrous and D. Touchard et al., Changes in the work of breathing induced by tracheotomy in ventilator-dependent patients, American Journal of Respiratory and Critical Care Medicine 159 (1999), pp. 383–388. View Record in Scopus | Cited By in Scopus (78)
34 A. Nieszkowska, A. Combes and C.E. Luyt et al., Impact of tracheotomy on sedative administration, sedation level, and comfort of mechanically ventilated intensive care unit patients, Critical Care Medicine 33 (2005), pp. 2527–2533. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (33)
35 M.J. Rumbak, M. Newton and T. Truncale et al., A prospective, randomized, study comparing early percutaneous dilational tracheotomy to prolonged translaryngeal intubation (delayed tracheotomy) in critically ill medical patients, Critical Care Medicine 32 (2004), pp. 1689–1694. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (133)
36 N. Hubner, W. Rees and K. Seufert et al., Percutaneous dilatational tracheostomy done early after cardiac surgery–outcome and incidence of mediastinitis, The Thoracic and Cardiovascular Surgeon 46 (1998), pp. 89–92. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (25)
*37 M.K. Banbury, M.E. Brizzio and J. Rajeswaran et al., Transfusion increases the risk of postoperative infection after cardiovascular surgery, Journal of the American College of Surgeons 202 (2006), pp. 131–138. Article | PDF (406 K) | View Record in Scopus | Cited By in Scopus (35)
38 B. Soderquist, Surgical site infections in cardiac surgery: microbiology, APMIS 115 (2007), pp. 1008–1011. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (4)
39 A.G. Gristina, P. Naylor and Q. Myrvik, Infections from biomaterials and implants: a race for the surface, Medical Progress Through Technology 14 (1988), pp. 205–224. View Record in Scopus | Cited By in Scopus (54)
40 D.R. Reagan, B.N. Doebbeling and M.A. Pfaller et al., Elimination of coincident Staphylococcus aureus nasal and hand carriage with intranasal application of mupirocin calcium ointment, Annals of Internal Medicine 114 (1991), pp. 101–106. View Record in Scopus | Cited By in Scopus (151)
41 H.J. Weinstein, The relation between the nasal-staphylococcal-carrier state and the incidence of postoperative complications, The New England Journal of Medicine 260 (1959), pp. 1303–1308.
42 R.E. Williams, M.P. Jevons and R.A. Shooter et al., Nasal staphylococci and sepsis in hospital patients, British Medical Journal 2 (1959), pp. 658–662. Full Text via CrossRef
43 X. Corbella, M.A. Dominguez and M. Pujol et al., Staphylococcus aureus nasal carriage as a marker for subsequent staphylococcal infections in intensive care unit patients, European Journal of Clinical Microbiology & Infectious Diseases 16 (1997), pp. 351–357. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (50)
44 H.G. Jakob, M. Borneff-Lipp and A. Bach et al., The endogenous pathway is a major route for deep sternal wound infection, European Journal of Cardio-Thoracic Surgery 17 (2000), pp. 154–160. Article | PDF (175 K) | View Record in Scopus | Cited By in Scopus (44)
45 J.A. Kluytmans, J.W. Mouton and E.P. Ijzerman et al., Nasal carriage of Staphylococcus aureus as a major risk factor for wound infections after cardiac surgery, The Journal of Infectious Diseases 171 (1995), pp. 216–219. View Record in Scopus | Cited By in Scopus (149)
46 S.E. Capes, D. Hunt and K. Malmberg et al., Stress hyperglycaemia and increased risk of death after myocardial infarction in patients with and without diabetes: a systematic overview, Lancet 355 (2000), pp. 773–778. Article | PDF (91 K) | View Record in Scopus | Cited By in Scopus (674)
*47 G. van den Berghe, P. Wouters and F. Weekers et al., Intensive insulin therapy in the critically ill patients, The New England Journal of Medicine 345 (2001), pp. 1359–1367. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (3319)
*48 A.P. Furnary, K.J. Zerr and G.L. Grunkemeier et al., Continuous intravenous insulin infusion reduces the incidence of deep sternal wound infection in diabetic patients after cardiac surgical procedures, Annals of Thoracic Surgery 67 (1999), pp. 352–360 discussion 60–2. Abstract | Article | PDF (250 K)
*49 K.J. Zerr, A.P. Furnary and G.L. Grunkemeier et al., Glucose control lowers the risk of wound infection in diabetics after open heart operations, Annals of Thoracic Surgery 63 (1997), pp. 356–361. Abstract | Article | PDF (406 K) | View Record in Scopus | Cited By in Scopus (325)
*50 G.Y. Gandhi, G.A. Nuttall and M.D. Abel et al., Intensive intraoperative insulin therapy versus conventional glucose management during cardiac surgery: a randomized trial, Annals of Internal Medicine 146 (2007), pp. 233–243. View Record in Scopus | Cited By in Scopus (100)
51 I.R. Hudson, The efficacy of intranasal mupirocin in the prevention of staphylococcal infections: a review of recent experience, The Journal of Hospital Infection 27 (1994), pp. 81–98. Abstract | Article | PDF (1473 K) | View Record in Scopus | Cited By in Scopus (82)
52 T.M. Perl and J.E. Golub, New approaches to reduce Staphylococcus aureus nosocomial infection rates: treating S. aureus nasal carriage, Annals of Pharmacotherapy 32 (1998), pp. S7–S16. View Record in Scopus | Cited By in Scopus (63)
53 G.E. Cimochowski, M.D. Harostock and R. Brown et al., Intranasal mupirocin reduces sternal wound infection after open heart surgery in diabetics and nondiabetics, Annals of Thoracic Surgery 71 (2001), pp. 1572–1578 discussion 8–9.
54 J.A. Kluytmans, J.W. Mouton and M.F. VandenBergh et al., Reduction of surgical-site infections in cardiothoracic surgery by elimination of nasal carriage of Staphylococcus aureus, Infection Control and Hospital Epidemiology 17 (1996), pp. 780–785. View Record in Scopus | Cited By in Scopus (129)
55 A. Konvalinka, L. Errett and I.W. Fong, Impact of treating Staphylococcus aureus nasal carriers on wound infections in cardiac surgery, The Journal of Hospital Infection 64 (2006), pp. 162–168. Article | PDF (120 K) | View Record in Scopus | Cited By in Scopus (15)
56 C. Pena, N. Fernandez-Sabe and M.A. Dominguez et al., Staphylococcus aureus nasal carriage in patients on haemodialysis: role of cutaneous colonization, The Journal of Hospital Infection 58 (2004), pp. 20–27. Abstract | PDF (211 K) | View Record in Scopus | Cited By in Scopus (9)
57 M. Perez-Fontan, M. Rosales and A. Rodriguez-Carmona et al., Mupirocin resistance after long-term use for Staphylococcus aureus colonization in patients undergoing chronic peritoneal dialysis, American Journal of Kidney Diseases 39 (2002), pp. 337–341. Abstract | PDF (38 K) | View Record in Scopus | Cited By in Scopus (56)
58 T.M. Perl, J.J. Cullen and R.P. Wenzel et al., Intranasal mupirocin to prevent postoperative Staphylococcus aureus infections, The New England Journal of Medicine 346 (2002), pp. 1871–1877. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (208)
59 A.B. Kaiser, D.S. Kernodle and N.L. Barg et al., Influence of preoperative showers on staphylococcal skin colonization: a comparative trial of antiseptic skin cleansers, Annals of Thoracic Surgery 45 (1988), pp. 35–38. View Record in Scopus | Cited By in Scopus (30)
60 N.A. Byrne Dj and A. Cuschieri, The value of whole body disinfection in the prevention of post-operative wound infection in clean and potentially contaminated surgery, Surgical Research Communications 12 (1992), pp. 43–52.
61 J. Webster and S. Osborne, Preoperative bathing or showering with skin antiseptics to prevent surgical site infection, Cochrane Database of Systematic Review (2007) CD004985.
62 W.A. Sampathkumar P, J.M. Manahan, C.J. Mullany and R.L. Thompson, Results of an Infection Control Program aimed at reducing Surgical Site Infections after Cardiac Surgery, Clinical Infectious Diseases 33 (2001) Abstract #240.
63 L.M. Brady, M. Thomson and M.A. Palmer et al., Successful control of endemic MRSA in a cardiothoracic surgical unit, Medical Journal of Australia 152 (1990), pp. 240–245. View Record in Scopus | Cited By in Scopus (28)
64 D.W. Bratzler and P.M. Houck, Antimicrobial prophylaxis for surgery: an advisory statement from the National Surgical Infection Prevention Project, Clinical Infectious Diseases 38 (2004), pp. 1706–1715. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (185)
65 A.J. Mangram, T.C. Horan and M.L. Pearson et al., Guideline for prevention of surgical site infection, 1999. Hospital Infection Control Practices Advisory Committee, Infection Control and Hospital Epidemiology 20 (1999), pp. 250–278 quiz 79–80. View Record in Scopus | Cited By in Scopus (443)
*66 R. Finkelstein, G. Rabino and T. Mashiah et al., Vancomycin versus cefazolin prophylaxis for cardiac surgery in the setting of a high prevalence of methicillin-resistant staphylococcal infections, The Journal of Thoracic and Cardiovascular Surgery 123 (2002), pp. 326–332. Abstract | PDF (116 K) | View Record in Scopus | Cited By in Scopus (53)
*67 M.K. Bolon, M. Morlote and S.G. Weber et al., Glycopeptides are no more effective than beta-lactam agents for prevention of surgical site infection after cardiac surgery: a meta-analysis, Clinical Infectious Diseases 38 (2004), pp. 1357–1363. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (25)
68 A.M. Eklund, Prevention of sternal wound infections with locally administered gentamicin, APMIS 115 (2007), pp. 1022–1024. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
69 O. Friberg, Local collagen-gentamicin for prevention of sternal wound infections: the LOGIP trial, APMIS 115 (2007), pp. 1016–1021. Full Text via CrossRef | View Record in Scopus | Cited By in Scopus (2)
  相似文献   

11.
Since the introduction of the REV procedure in 1980, this operation has become our procedure of choice for patients with transposition of the great arteries, VSD, and POT obstruction. The classical Rastelli procedure is indicated only in patients in whom the presence of a well-functioning pulmonary valve is deemed necessary (i.e., hypoplastic right ventricle, inadequate pulmonary arterial bed). Compared with the Rastelli operation, the REV procedure may have definite advantages:
1. The REV operation creates a straight, short left ventricular-aortic tunnel, provides proper alignment of the aorta with the left ventricular chamber, and practically eliminates the risk of late subaortic stenosis.
2. Complete repair is feasible in patients in whom a Rastelli procedure would be contraindicated because of unfavorable intracardiac anatomy (i.e., restrictive VSD, abnormal attachment of tricuspid chordae on the conal septum).
3. The absence of an extracardiac conduit to restore the right ventricular-pulmonary artery continuity reduces the need for reoperation due to recurrent POT obstruction. In most patients, the long-term outcome of the reconstructed right ventricular outflow tract may be similar to that of a repaired tetralogy of Fallot. However, the need to relieve right ventricular outflow tract obstruction, although reduced in comparison with the Rastelli procedure, is not eliminated completely.
4. Because of its feasibility in most intracardiac situations and because it obviates the need for an extracardiac conduit, the REV procedure can be performed in infants and small children, thus avoiding, in most patients, an initial palliative step.

References

1 Y Lecompte, A Batisse and D Di Carlo, Double-outlet right ventricle: A surgical synthesis, in (ed), Advances in Cardiac Surgery vol 4, Mosby Year Book (1993), pp. 109–136. View Record in Scopus | Cited By in Scopus (5)
2 R Prêtre, G Gendron and D Tamisier et al., Results of the Lecompte procedure in malposition of the great arteries and pulmonary obstruction, Eur J Cardiothorac Surg 19 (2001), pp. 283–289. Article | PDF (214 K) | View Record in Scopus | Cited By in Scopus (6)
3 MW Turrentine, RP McCarthy and P Vijay et al., PTFE monocusp valve reconstruction of the right ventricular outflow tract, Ann Thorac Surg 73 (2002), pp. 871–879.
  相似文献   

12.
13.
14.
15.
16.
17.
18.
19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号