Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Richard F. Jacobs is active.

Publication


Featured researches published by Richard F. Jacobs.


The Journal of Pediatrics | 2003

Effect of ganciclovir therapy on hearing in symptomatic congenital cytomegalovirus disease involving the central nervous system: a randomized, controlled trial∗

David W. Kimberlin; Chin-Yu Lin; Pablo J. Sánchez; Gail J. Demmler; Wayne M. Dankner; Mark J. Shelton; Richard F. Jacobs; Wendy Vaudry; Robert F. Pass; Jan Kiell; Seng-jaw Soong; Richard J. Whitley

OBJECTIVE To evaluate the efficacy and safety of ganciclovir therapy in neonates with congenital cytomegalovirus (CMV) disease. STUDY DESIGN Neonates with symptomatic CMV disease involving the central nervous system were randomly assigned to receive 6 weeks of intravenous ganciclovir versus no treatment. The primary end point was improved brainstem-evoked response (BSER) between baseline and 6-month follow-up (or, for patients with normal baseline hearing, normal BSER at both time points). RESULTS From 1991 to 1999, 100 patients were enrolled. Of these, 42 patients had both a baseline and 6-month follow-up BSER audiometric examination and thus were evaluable for the primary end point. Twenty-one (84%) of 25 ganciclovir recipients had improved hearing or maintained normal hearing between baseline and 6 months versus 10 (59%) of 17 control patients (P=.06). None (0%) of 25 ganciclovir recipients had worsening in hearing between baseline and 6 months versus 7 (41%) of 17 control patients (P<.01). A total of 43 patients had a BSER at both baseline and at 1 year or beyond. Five (21%) of 24 ganciclovir recipients had worsening of hearing between baseline and > or =1 year versus 13 (68%) of 19 control patients (P<.01). A total of 89 patients had absolute neutrophil counts determined during the course of the study; 29 (63%) of 46 ganciclovir-treated patients had grade 3 or 4 neutropenia during treatment versus 9 (21%) of 43 control patients (P<.01). CONCLUSIONS Ganciclovir therapy begun in the neonatal period in symptomatically infected infants with CMV infection involving the central nervous system prevents hearing deterioration at 6 months and may prevent hearing deterioration at > or =1 year. Almost two thirds of treated infants have significant neutropenia during therapy.


The New England Journal of Medicine | 1991

Predictors of Morbidity and Mortality in Neonates with Herpes Simplex Virus Infections

Richard J. Whitley; Ann M. Arvin; Charles G. Prober; Lawrence Corey; Sandra K. Burchett; Stanley A. Plotkin; Stuart E. Starr; Richard F. Jacobs; Dwight A. Powell; Andre J. Nahmias; Ciro V. Sumaya; Kathryn M. Edwards; Charles A. Alford; Gary Caddell; Seng-jaw Soong

BACKGROUND In a controlled trial comparing acyclovir with vidarabine in the treatment of neonatal herpes simplex virus (HSV) infection, we found no significant difference between the treatments in adjusted mortality and morbidity. Hence, we sought to define for the entire cohort (n = 202) the clinical characteristics that best predicted the eventual outcome in these neonates. METHODS Data were gathered prospectively at 27 centers between 1981 and 1988 in infants less than one month of age who had virologically confirmed HSV infection. We examined the outcomes by multivariate analyses of 24 variables. Disease was classified in one of three categories based on the extent of the involvement at entry into the trial: infection confined to skin, eyes, or mouth; encephalitis; or disseminated infection. RESULTS AND CONCLUSIONS There were no deaths among the 85 infants with localized HSV infection. The mortality rate was significantly higher in the 46 neonates with disseminated infection (57 percent) than in the 71 with encephalitis (15 percent). In addition, the risk of death was increased in neonates who were in or near coma at entry (relative risk, 5.2), had disseminated intravascular coagulopathy (relative risk, 3.8), or were premature (relative risk, 3.7). In babies with disseminated disease, HSV pneumonitis was also associated with greater mortality (relative risk, 3.6). In the survivors, morbidity was most frequent in infants with encephalitis (relative risk, 4.4), disseminated infection (relative risk, 2.1), seizures (relative risk, 3.0), or infection with HSV type 2 (relative risk, 4.9). With HSV infection limited to the skin, eyes, or mouth, the presence of three or more recurrences of vesicles was associated with an increased risk of neurologic impairment as compared with two or fewer recurrences.


Pediatrics | 1998

Prevention of respiratory syncytial virus infections: Indications for the use of palivizumab and update on the use of RSV-IGIV

Neal A. Halsey; Jon S. Abramson; P. Joan Chesney; Margaret C. Fisher; Michael A. Gerber; S. Michael Marcy; Dennis L. Murray; Gary D. Overturf; Charles G. Prober; Thomas N. Saari; Leonard B. Weiner; Richard J. Whitley; R. Breiman; M. Carolyn Hardegree; A. Hirsch; Richard F. Jacobs; N. E. MacDonald; Walter A. Orenstein; N. Regina Rabinovich; B. Schwartz; Georges Peter; Carol J. Baker; Larry K. Pickering; H. Cody Meissner; James A. Lemons; Lillian R. Blackmon; William P. Kanto; Hugh MacDonald; Carol Miller; Lu Ann Papile

The Food and Drug Administration recently approved the use of palivizumab (palē-vizhū-mäb), an intramuscularly administered monoclonal antibody preparation. Recommendations for its use are based on a large, randomized study demonstrating a 55% reduction in the risk of hospitalization attributable to respiratory syncytial virus (RSV) infections in high-risk pediatric patients. Infants and children with chronic lung disease (CLD), formerly designated bronchopulmonary dysplasia, as well as prematurely born infants without CLD experienced a reduced number of hospitalizations while receiving palivizumab compared with a placebo. Both palivizumab and respiratory syncytial virus immune globulin intravenous (RSV-IGIV) are available for protecting high-risk children against serious complications from RSV infections. Palivizumab is preferred for most high-risk children because of ease of administration (intramuscular), lack of interference with measles–mumps–rubella vaccine and varicella vaccine, and lack of complications associated with intravenous administration of human immune globulin products. RSV-IGIV, however, provides additional protection against other respiratory viral illnesses and may be preferred for selected high-risk children including those receiving replacement intravenous immune globulin because of underlying immune deficiency or human immuno-deficiency virus infection. For premature infants about to be discharged from hospitals during the RSV season, physicians could consider administering RSV-IGIV for the first month of prophylaxis. Most of the guidelines from the American Academy of Pediatrics for the selection of infants and children to receive RSV-prophylaxis remain unchanged. Palivizumab has been shown to provide benefit for infants who were 32 to 35 weeks of gestation at birth. RSV-IGIV is contraindicated and palivizumab is not recommended for children with cyanotic congenital heart disease. The number of patients with adverse events judged to be related to palivizumab was similar to that of the placebo group (11% vs 10%, respectively); discontinuation of injections for adverse events related to palivizumab was rare.


The New England Journal of Medicine | 1991

A CONTROLLED TRIAL COMPARING VIDARABINE WITH ACYCLOVIR IN NEONATAL HERPES SIMPLEX VIRUS INFECTION

Richard J. Whitley; Ann M. Arvin; Charles G. Prober; Sandra K. Burchett; Lawrence Corey; Dwight A. Powell; Stanley A. Plotkin; Stuart E. Starr; Charles A. Alford; James D. Connor; Richard F. Jacobs; Andre J. Nahmias; Seng-Jaw Soong

BACKGROUND Despite the use of vidarabine, herpes simplex virus (HSV) infection in neonates continues to be a disease of high morbidity and mortality. We undertook a controlled trial comparing vidarabine with acyclovir for the treatment of neonatal HSV infection. METHODS Babies less than one month of age with virologically confirmed HSV infection were randomly and blindly assigned to receive either intravenous vidarabine (30 mg per kilogram of body weight per day; n = 95) or acyclovir (30 mg per kilogram per day; n = 107) for 10 days. Actuarial rates of mortality and morbidity among the survivors after one year were compared overall and according to the extent of the disease at entry into the study (infection confined to the skin, eyes, or mouth; encephalitis; or disseminated disease). RESULTS After adjustment for differences between groups in the extent of disease, there was no difference between vidarabine and acyclovir in either morbidity (P = 0.83) or mortality (P = 0.27). None of the 85 babies with disease confined to the skin, eyes, or mouth died. Of the 31 babies in this group who were treated with vidarabine and followed for a year, 88 percent (22 of 25) were judged to be developing normally after one year, as compared with 98 percent (45 of 46) of the 54 treated with acyclovir (95 percent confidence interval for the difference, -4 to 24). For the 71 babies with encephalitis, mortality was 14 percent with vidarabine (5 of 36) and with acyclovir (5 of 35); of the survivors, 43 percent (13 of 30) and 29 percent (8 of 28), respectively, were developing normally after one year (95 percent confidence interval for the difference, -11 to 39). For the 46 babies with disseminated disease, mortality was 50 percent (14 of 28) with vidarabine and 61 percent (11 of 18) with acyclovir (95 percent confidence interval for the difference, -20 to 40); of the survivors, 58 percent (7 of 12) and 60 percent (3 of 5), respectively, were judged to be developing normally after one year (95 percent confidence interval for the difference, -40 to 50). Both medications were without serious toxic effects. CONCLUSIONS In this multicenter, randomized, blinded study there were no differences in outcome between vidarabine and acyclovir in the treatment of neonatal HSV infection. The study lacked statistical power to determine whether there were sizable differences within the subgroups of those with localized HSV, encephalitis, or disseminated disease.


The Journal of Infectious Diseases | 1997

Ganciclovir Treatment of Symptomatic Congenital Cytomegalovirus Infection: Results of a Phase II Study

Richard J. Whitley; Gretchen A. Cloud; William C. Gruber; Gregory A. Storch; Gail J. Demmler; Richard F. Jacobs; Wayne M. Dankner; Stephen A. Spector; Stuart E. Starr; Robert F. Pass; Sergio Stagno; William J. Britt; Charles A. Alford; Seng-jaw Soong; Xiao-Jian Zhou; Lanette Sherrill; Jan M. FitzGerald; Jean-Pierre Sommadossi

Congenital cytomegalovirus (CMV) infection occurs in approximately 1% of newborns in the United States. A phase II evaluation was done of ganciclovir for the treatment of symptomatic congenital CMV infection. Daily doses of 8 or 12 mg/kg were administered in divided doses at 12-h intervals for 6 weeks. Clinical and laboratory evaluations sought evidence of toxicity, quantitative virologic responses in urine, plasma drug concentrations, and clinical outcome. A total of 14 and 28 babies received 8 and 12 mg/kg/day, respectively. Five additional babies received ganciclovir on a compassionate plea basis. Significant laboratory abnormalities included thrombocytopenia (< or = 50,000/mm3) in 37 babies and absolute neutropenia (< or = 500 mm3) in 29 babies. Quantitative excretion of CMV in the urine decreased; however, after cessation of therapy, viruria returned to near pretreatment levels. Hearing improvement or stabilization occurred in 5 (16%) of 30 babies at 6 months or later, indicating efficacy.


The Journal of Pediatrics | 1987

Intrauterine herpes simplex virus infections

Cecelia Hutto; Ann M. Arvin; Richard F. Jacobs; Russell W. Steele; Sergio Stagno; Raymond Lyrene; Lynne D. Willett; Dwight A. Powell; Richard Andersen; Joe Werthammer; Gilbert Ratcliff; Andre J. Nahmias; Cynthia Christy; Richard J. Whitley

Neonatal herpes simplex virus (HSV) infection is usually acquired at birth, although a few infants have had findings suggestive of intrauterine infection. We describe 13 babies who had clinical manifestations of intrauterine HSV infection, including skin lesions and scars at birth (12), chorioretinitis (eight), microcephaly (seven), hydranencephaly (five), and microphthalmia (two). All infants had combinations of these defects. Infection was proved by viral isolation in each case; all isolates were HSV-2. Two infants died during the first week of life; 10 of the surviving infants had severe neurologic sequelae, and one infant was blind. Four mothers experienced an apparent primary genital HSV infection, and one had recurrent infection, at varying times during gestation. The remaining women denied a history of symptoms of genital HSV infection. These findings indicate that intrauterine HSV infection can occur as a consequence of either primary or recurrent maternal infection and has severe consequences for the fetus.


The Journal of Infectious Diseases | 2008

Pharmacokinetic and Pharmacodynamic Assessment of Oral Valganciclovir in the Treatment of Symptomatic Congenital Cytomegalovirus Disease

David W. Kimberlin; Edward P. Acosta; Pablo J. Sánchez; Sunil K. Sood; Vish Agrawal; James Homans; Richard F. Jacobs; David Lang; Jose R. Romero; Jill Griffin; Gretchen A. Cloud; Fred D. Lakeman; Richard J. Whitley

BACKGROUND Intravenous ganciclovir administered for 6 weeks improves hearing outcomes in infants with symptomatic congenital cytomegalovirus (CMV) disease involving the central nervous system. METHODS Twenty-four subjects received antiviral therapy for 6 weeks. Serial pharmacokinetic assessments were performed after administration of valganciclovir oral solution and of intravenous ganciclovir. RESULTS On the basis of a previous pharmacokinetic study of the use of intravenous ganciclovir in this population, a target AUC12 (area under the concentration-time curve over a 12-h period) of 27 mg x h/L was defined. The median dose of oral valganciclovir administered in the present trial was 16 mg/kg, which produced a geometric mean AUC12 of 27.4 mg x h/L. The bioavailability of valganciclovir was 41.1%. Of the 18 subjects who had detectable CMV in whole blood at baseline or during therapy, 11 had <4 log viral DNA copies/mL at baseline, and 7 had > or =4 log viral DNA copies/mL at baseline; subjects who started the study with the higher viral burden experienced greater decreases in viral load but did not clear virus during the 42-day course of therapy. Neutropenia of grade 3 or 4 developed in 38% of subjects. CONCLUSIONS In neonates with symptomatic congenital CMV disease, valganciclovir oral solution provides plasma concentrations of ganciclovir comparable to those achieved with administration of intravenous ganciclovir. The results of the present study cannot be extrapolated to extemporaneously compounded liquid formulations of valganciclovir.


The New England Journal of Medicine | 2011

Oral Acyclovir Suppression and Neurodevelopment after Neonatal Herpes

David W. Kimberlin; Richard J. Whitley; Wen Wan; Dwight A. Powell; Gregory A. Storch; Amina Ahmed; April L. Palmer; Pablo J. Sánchez; Richard F. Jacobs; John S. Bradley; Joan Robinson; Mark J. Shelton; Penelope H. Dennehy; Charles T. Leach; Mobeen H. Rathore; Nazha Abughali; Peter F. Wright; Lisa M. Frenkel; Rebecca C. Brady; Russell B. Van Dyke; Leonard B. Weiner; Judith Guzman-Cottrill; Carol A. McCarthy; Jill Griffin; Penelope Jester; Misty Parker; Fred D. Lakeman; Huichien Kuo; Choo Hyung Lee; Gretchen A. Cloud

BACKGROUND Poor neurodevelopmental outcomes and recurrences of cutaneous lesions remain unacceptably frequent among survivors of neonatal herpes simplex virus (HSV) disease. METHODS We enrolled neonates with HSV disease in two parallel, identical, double-blind, placebo-controlled studies. Neonates with central nervous system (CNS) involvement were enrolled in one study, and neonates with skin, eye, and mouth involvement only were enrolled in the other. After completing a regimen of 14 to 21 days of parenteral acyclovir, the infants were randomly assigned to immediate acyclovir suppression (300 mg per square meter of body-surface area per dose orally, three times daily for 6 months) or placebo. Cutaneous recurrences were treated with open-label episodic therapy. RESULTS A total of 74 neonates were enrolled--45 with CNS involvement and 29 with skin, eye, and mouth disease. The Mental Development Index of the Bayley Scales of Infant Development (in which scores range from 50 to 150, with a mean of 100 and with higher scores indicating better neurodevelopmental outcomes) was assessed in 28 of the 45 infants with CNS involvement (62%) at 12 months of age. After adjustment for covariates, infants with CNS involvement who had been randomly assigned to acyclovir suppression had significantly higher mean Bayley mental-development scores at 12 months than did infants randomly assigned to placebo (88.24 vs. 68.12, P=0.046). Overall, there was a trend toward more neutropenia in the acyclovir group than in the placebo group (P=0.09). CONCLUSIONS Infants surviving neonatal HSV disease with CNS involvement had improved neurodevelopmental outcomes when they received suppressive therapy with oral acyclovir for 6 months. (Funded by the National Institute of Allergy and Infectious Diseases; CASG 103 and CASG 104 ClinicalTrials.gov numbers, NCT00031460 and NCT00031447, respectively.).


Clinical Infectious Diseases | 1998

Bartonella henselae as a Cause of Prolonged Fever and Fever of Unknown Origin in Children

Richard F. Jacobs; Gordon E. Schutze

A prospective evaluation of 146 children with fever of unknown origin (FUO) and prolonged fever was performed from 1990 to 1996. FUO was defined as a documented daily temperature of > or = 38 degrees C for at least 14 days without diagnostic signs or symptoms. Prolonged fever was defined as fever for at least 14 days and no diagnosis at the time of referral for evaluation. An established diagnosis was made for 84 (57.5%) of 146 patients. The most common infectious disease diagnoses were Epstein-Barr virus infection (22 [15.1%] of 146), osteomyelitis (14 [9.6%] of 146), bartonellosis (7 [4.8%] of 146), and urinary tract infection (6 [4.1%] of 146). Three of seven patients with confirmed Bartonella henselae infection presented with FUO and no ultrasonographic findings compatible with hepatosplenic involvement; two patients presented with FUO and hepatosplenic involvement. The relatively common finding of acute bartonellosis in this population suggests that FUO and prolonged fever in children are other presentations of infection with B. henselae.


Pediatric Infectious Disease Journal | 1996

Administration of oral acyclovir suppressive therapy after neonatal herpes simplex virus disease limited to the skin, eyes and mouth: Results of a phase I/II trial

David W. Kimberlin; Dwight A. Powell; William C. Gruber; Pamela S. Diaz; Ann M. Arvin; Mary L. Kumar; Richard F. Jacobs; Russell B. Van Dyke; Sandra K. Burchett; Seng Jaw Soong; Alfred D. Lakeman; Richard J. Whitley; C. Laughlin; Richard Whitley; A. Lakeman; S. J. Soong; D. Kimberlin; Sergio Stagno; Robert F. Pass; A. Arvin; Charles G. Prober; John S. Bradley; Stephen A. Spector; Larry Corey; Gail J. Demmler; S. Burchett; Stuart P. Adler; James F. Bale; Yvonne J. Bryson; Tasnee Chonmaitree

BACKGROUND Neonatal herpes simplex virus (HSV) infections limited to the skin, eyes and mouth (SEM) can result in neurologic impairment. A direct correlation exists between the development of neurologic deficits and the frequency of cutaneous HSV recurrences. Thus, the National Institutes of Allergy and Infectious Diseases Collaborative Antiviral Study Group conducted a Phase I/II trial of oral acyclovir therapy for the suppression of cutaneous recurrences after SEM disease in 26 neonates. METHODS Infants < or = 1 month of age with virologically confirmed HSV-2 SEM disease were eligible for enrollment. Suppressive oral acyclovir therapy (300 mg/m2/dose given either twice daily or three times per day) was administered for 6 months. RESULTS Twelve (46%) of the 26 infants developed neutropenia (< 1000 cells/mm3) while receiving acyclovir. Thirteen (81%) of the 16 infants who received drug 3 times per day experienced no recurrences of skin lesions while receiving therapy. In comparison, a previous Collaborative Antiviral Study Group study found that only 54% of infants have no cutaneous recurrences in the 6 months after resolution of neonatal HSV disease if oral acyclovir suppressive therapy is not initiated. In one infant, HSV DNA was detected in the cerebrospinal fluid during a cutaneous recurrence, and an acyclovir-resistant HSV mutant was isolated from another patient during the course of the study. CONCLUSIONS Administration of oral acyclovir can prevent cutaneous recurrences of HSV after neonatal SEM disease. The effect of such therapy on neurologic outcome must be assessed in a larger, Phase III study. As such, additional investigation is necessary before routine use of suppressive therapy in this population can be recommended.

Collaboration


Dive into the Richard F. Jacobs's collaboration.

Top Co-Authors

Avatar

Richard J. Whitley

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Gregory L. Kearns

Arkansas Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Gordon E. Schutze

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

David W. Kimberlin

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Neal A. Halsey

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

R. Yogev

Children's Memorial Hospital

View shared research outputs
Top Co-Authors

Avatar

Dennis L. Murray

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge