You are on page 1of 48

Small Animal Surgical Emergencies

2nd Edition Lillian R. Aronson


Visit to download the full and correct content document:
https://ebookmass.com/product/small-animal-surgical-emergencies-2nd-edition-lillian-
r-aronson/
Small Animal Surgical Emergencies
Small Animal Surgical Emergencies

Second Edition

Edited by

Lillian R. Aronson
Professor of Surgery
School of Veterinary Medicine
University of Pennsylvania
Philadelphia, PA, USA
This second edition first published 2022
© 2022 John Wiley & Sons, Inc.

Edition History
John Wiley & Sons, Inc. (1e, 2016)

All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means,
electronic, mechanical, photocopying, recording or otherwise, except as permitted by law. Advice on how to obtain permission to reuse material
from this title is available at http://www.wiley.com/go/permissions.

The right of Lillian R. Aronson to be identified as the author of the editorial material in this work has been asserted in accordance with law.

Registered Office
John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, USA

Editorial Office
111 River Street, Hoboken, NJ 07030, USA

For details of our global editorial offices, customer services, and more information about Wiley products visit us at www.wiley.com.

Wiley also publishes its books in a variety of electronic formats and by print-­on-­demand. Some content that appears in standard print versions
of this book may not be available in other formats.

Limit of Liability/Disclaimer of Warranty


The contents of this work are intended to further general scientific research, understanding, and discussion only and are not intended and
should not be relied upon as recommending or promoting scientific method, diagnosis, or treatment by physicians for any particular patient.
In view of ongoing research, equipment modifications, changes in governmental regulations, and the constant flow of information relating
to the use of medicines, equipment, and devices, the reader is urged to review and evaluate the information provided in the package insert
or instructions for each medicine, equipment, or device for, among other things, any changes in the instructions or indication of usage and
for added warnings and precautions. While the publisher and authors have used their best efforts in preparing this work, they make no
representations or warranties with respect to the accuracy or completeness of the contents of this work and specifically disclaim all warranties,
including without limitation any implied warranties of merchantability or fitness for a particular purpose. No warranty may be created or
extended by sales representatives, written sales materials or promotional statements for this work. The fact that an organization, website, or
product is referred to in this work as a citation and/or potential source of further information does not mean that the publisher and authors
endorse the information or services the organization, website, or product may provide or recommendations it may make. This work is sold with
the understanding that the publisher is not engaged in rendering professional services. The advice and strategies contained herein may not be
suitable for your situation. You should consult with a specialist where appropriate. Further, readers should be aware that websites listed in this
work may have changed or disappeared between when this work was written and when it is read. Neither the publisher nor authors shall be
liable for any loss of profit or any other commercial damages, including but not limited to special, incidental, consequential, or other damages.

Library of Congress Cataloging-­in-­Publication Data


Names: Aronson, Lillian R., editor.
Title: Small animal surgical emergencies / edited by Lillian R. Aronson.
Description: Second edition. | Hoboken, NJ : Wiley-Blackwell, 2022. |
Includes bibliographical references and index.
Identifiers: LCCN 2022000548 (print) | LCCN 2022000549 (ebook) | ISBN
9781119658535 (cloth) | ISBN 9781119658610 (adobe pdf) | ISBN
9781119658627 (epub)
Subjects: MESH: Surgery, Veterinary–methods | Pets–surgery |
Emergencies–veterinary | Animal Diseases–surgery | Wounds and
Injuries–veterinary
Classification: LCC SF914.3 (print) | LCC SF914.3 (ebook) | NLM SF 914.3
| DDC 636.089/7–dc23/eng/20220209
LC record available at https://lccn.loc.gov/2022000548
LC ebook record available at https://lccn.loc.gov/2022000549

Cover Design: Wiley


Cover Images: Courtesy of Hunter Piegols, Courtesy of Marie Burneko

Set in 9.5/12.5pt STIXTwoText by Straive, Pondicherry, India

10 9 8 7 6 5 4 3 2 1
March 3, 1963

This textbook is dedicated to my amazing parents, Raphael and Dorothea Aronson. Growing up, my parents created a wel-
coming home filled with warmth, love, and constant laughter. They stressed the importance of family and community and
have always been incredible role models, instilling in me the importance of honesty, respect, to always remain humble and
to do the right thing even when it is not popular. From a very young age, my parents supported and encouraged me to
follow my passion for veterinary medicine. I wouldn’t be where I am today if it wasn’t for their love and support. I consider
myself the luckiest girl in the world, to be their daughter.
vii

Contents

Contributors xii
Preface xvii
Acknowledgements xviii
About the Companion Website xix
Daisy’s Story xx

1 Triage and Initial Stabilization of the Emergency Small Animal Surgical Patient 1
Dana Clarke

2 Operating Room Nursing Tips for Emergency Surgical Procedures 20


Cami Elliott, Michelle Capps, and Michael McCallum

Section I Gastrointestinal System 49

3 Emergency Stabilization of the Acute Abdomen Patient 51


Nyssa Levy and Matthew Beal

4 Esophageal Foreign Bodies 58


Ivan Doran

5 Gastrointestinal Foreign Bodies 70


Amie Koenig and Mandy L. Wallace

6 Intussusception 81
Janet Kovak McClaran and Yekaterina Buriko

7 Rectal Prolapse 88
Jennifer L. Huck

8 Gastric Dilatation and Volvulus 93


Michael S. Tivers and Sophie Adamantos

9 Intestinal Volvulus 108


Elizabeth LaFond and Kristina Kiefer

10 Colonic Torsion 115


Chloe Wormser

11 Peritonitis 119
Adrienne Bentley and Anusha Balakrishnan
viii Contents

12 Surgical Intervention Post-­Celiotomy 141


Michael B. Mison

13 Biliary Emergencies 146


Lori Ludwig and Sean Smarick

14 Hemoperitoneum 164
Cassie N. Lux, William T. N. Culp and Steven E. Epstein

15 True Abdominal Hernias 178


Daniel D. Smeak

16 Traumatic and Incisional Hernias 190


Todd Hamilton

17 Perineal Hernias 199


Karen M. Tobias

18 Pancreatic Abscessation 216


F. A. (Tony) Mann

19 Hepatic Emergencies 226


James A. Perry, Steven E. Epstein, and William T. N. Culp

20 Bleeding Gastrointestinal Ulcers 238


Yekaterina Buriko, David Holt, Laura Ateca, and Lesley King

21 Enteral Feeding Tubes 251


Aarti Kathrani

Section II Urinary System 263

22 Stabilization of the Uremic Patient 265


Kari Beer and Kenneth Drobatz

Section IIa Urinary System - Urinary Tract Trauma 277

23 Renal and Ureteral Injury 279


Dan Degner and Chloe Wormser

24 Approach to the Uroabdomen 285


Sara Colopy, Adam Townsend, and Dale Bjorling

25 Urethral Injury 297


Sara Colopy, Adam Townsend, and Dale Bjorling

Section IIb Urinary System – Urinary Obstruction 311

26 Ureteral Obstruction 313


Daniel Degner, Dana Clarke, and Chloe Wormser

27 Urethral Obstruction 331


Heidi Phillips and Sara Colopy
Contents ix

Section III Respiratory System 351

Section IIIa Respiratory System – Upper Airway Disease 353

28 Stabilization Techniques for Patients with an Upper Respiratory Obstruction 355


Meredith Maczuzak and Deborah C. Silverstein

29 Tracheostomy 364
Nathaniel Lam

30 Brachycephalic Airway Syndrome 372


Mike Schlicksup

31 Laryngeal Paralysis 383


Georga T. Karbe

32 Laryngeal Trauma 391


Eric Monnet

33 Tracheal Collapse 395


William T. N. Culp and Matthew D. O’Donnell

Section IIIb Respiratory System – Pleural Space Disease 405

34 Stabilization Techniques for Patients with Pleural Space Disease 407


Erica Reineke and Jen Savini

35 Diaphragmatic Hernia 417


MaryAnn Radlinsky

36 Pyothorax 425
Lori S. Waddell and David A. Puerto

37 Hemothorax 433
Chad W. Schmiedt and Benjamin M. Brainard

38 Pneumothorax 441
David A. Puerto and Lori S. Waddell

39 Lung Lobe Torsion 451


Jacob A. Rubin and Jaime Green

40 Surgical Approach to the Thoracic Cavity 459


David A. Puerto and Jacob A. Rubin

Section IIIc Respiratory System – Chest Wall Disease 467

41 Stabilization Techniques for Patients with Chest Wall Disease 469


Nolan Chalifoux and Deborah Silverstein

42 Pectus Excavatum 475


Krista N. Adamovich-­Rippe and William T. N. Culp
x Contents

43 Flail Chest 483


Eric Monnet

Section IV Cardiovascular System 487

44 Pacemaker Therapy 489


Caryn Reynolds and Katrine Saile

45 Pericardial Effusion 501


Augusta Pelosi and Amy Koenigshof

46 Open-­Chest Cardiopulmonary Resuscitation 514


Jasper Burke and Deborah Silverstein

Section V Reproductive System 525

47 Cesarean Section 527


Stephen D. Gilson

48 Pyometra 534
Jacqueline Davidson and Dorothy Black

49 Penile and Testicular Emergencies 541


Susan P. Gregory

50 Prostatic Abscessation 553


Richard A. S. White

51 Uterine and Vaginal Prolapse 565


Pieter Nelissen

Section VI Wound Management 573

52 Bite Wounds 575


David Holt and Vincent Thawley

53 Burn Injury 586


Caroline Garzotto and Dana Clarke

54 Penetrating Injury in the Dog and Cat 600


Steve J. Mehler

55 Necrotizing Fasciitis 613


Karol A. Mathews and Ameet Singh

56 Degloving and Shear Injuries 626


Bryden J. Stanley and Susan W. Volk

57 Skin Flaps and Grafts: Managing Complications 645


Michael B. Mison
Contents xi

58 Incisional Infections 652


Brandy A. Burgess

Section VII Ocular Emergencies 659

59 Ocular Emergencies 661


Deborah C. Mandell and Rosalie M. Atkins

Section VIII Oral Surgical Emergencies 685

60 Oral Surgical Emergencies 687


Alexander M. Reiter

Section IX Orthopedic Emergencies 701

61 Approach to the Septic Joint 703


Samuel P. Franklin

62 Surgical Management of Open Fractures 715


Kimberly Agnello

63 Traumatic Joint Luxation and Reduction 725


Anna Massie and Po-­Yen Chou

Index 739
xii

Contributors

Sophie Adamantos BVSc CertVA DACVECC DECVCC Adrienne Bentley DVM DACVS
MRCVS FHEA Staff Surgeon
Clinical Director Cornell University Veterinary Specialists
Paragon Veterinary Referrals Stamford, CT, USA
Wakefield, UK
Dale Bjorling DVM DACVS
Krista N. Adamovich-­Rippe DVM DACVS Professor of Small Animal Surgery
Associate Surgeon University of Wisconsin-­Madison
Central Texas Veterinary Specialty and Emergency Hospital School of Veterinary Medicine
Austin, TX, USA Madison, WI, USA

Kimberly Agnello DVM DACVS Dorothy Black DVM MPVM DACVECC


Associate Professor of Surgery Criticalist and Owner
University of Pennsylvania, School of Veterinary Medicine Rex Vet, Inc., San Mateo, CA, USA
Philadelphia, PA, USA
Ben Brainard VMD DACVAA DACVECC
Laura Ateca VMD DACVECC Professor of Small Animal Critical Care
Attending Criticalist Director of Clinical Research
Emergency and Critical Care Service, Guardian Veterinary Department of Small Animal Medicine and Surgery
Specialists University of Georgia, College of Veterinary Medicine
Brewster, NY, USA Athens, GA, USA

Rosalie M. Atkins DVM DACVO Brandy A. Burgess DVM MSc PhD DACVIM-­LA DACVPM
VCA Northwest Veterinary Specialists, Associate Professor, Director of Infection Control
Clackamas, OR, USA University of Georgia, College of Veterinary Medicine
Athens, GA, USA
Anusha Balakrishnan BVSc DACVECC
Triangle Veterinary Referral Hospital, Kate Buriko DVM DACVECC
Durham, NC, USA Assistant Professor, Emergency and Critical Care
University of Pennsylvania, School of Veterinary Medicine
Matthew Beal DVM DACVECC Philadelphia, PA, USA
Head of Interventional Radiology Services,
Head of Emergency and Critical Care Medical Services Jasper Burke VMD
Department of Small Animal Clinical Sciences Resident in Emergency and Critical Care
Michigan State University, College of Veterinary Medicine University of Pennsylvania, School of Veterinary Medicine
East Lansing, MI, USA Philadelphia, PA, USA

Kari Santoro Beer DVM DACVECC Michelle Capps CVT


Emergency and Critical Care Specialist Surgical Nursing Staff Supervisor
Oakland Veterinary Referral Services University of Pennsylvania, School of Veterinary Medicine
Bloomfield Hills, MI, USA Philadelphia, PA, USA
Contributors xiii

Nolan Chalifoux DVM Cami Elliot CVT


Resident in Emergency and Critical Care Nashville Veterinary Specialists and Animal Emergency
University of Pennsylvania, School of Veterinary Medicine Nashville, TN, USA
Philadelphia, PA, USA
Steven E. Epstein DVM DACVECC
Po-­Yen Chou BVM MVM DACVS Professor of Emergency and Critical Care
Assistant Professor of Small Animal Orthopedic Surgery Department of Surgical and Radiological Sciences
Department of Surgical and Radiological Sciences University of California-­Davis, School of Veterinary
University of California-­Davis, School of Veterinary Medicine
Medicine Davis, CA, USA
Davis, CA, USA
Samuel P. Franklin DVM MS PhD DACVS DACVSMR
Dana Clarke DVM DACVECC ACVS Founding Fellow, Minimally Invasive Surgery,
Assistant Professor, Interventional Radiology and Small Animal Orthopedics
Critical Care Kansas City Canine Orthopedics, Kansas City, KS, USA
University of Pennsylvania, School of Veterinary Medicine
Philadelphia, PA, USA Caroline Garzotto VMD DACVS
Staff Surgeon
Sara Colopy DVM, PhD, DACVS Mount Laurel Animal Hospital
Clinical Assistant Professor, Small Animal Surgery Mount Laurel, NJ, USA
University of Wisconsin, School of Veterinary Medicine
Stephen D. Gilson DVM DACVS
Madison WI, USA
ACVS Founding Fellow in Surgical Oncology
Indigo Veterinary Specialists
William T.N. Culp VMD DACVS
Phoenix, AZ, USA
Professor of Small Animal Surgery
Department of Surgical and Radiological Sciences
Jaime Green DVM, DACVS
University of California-­Davis, School of Veterinary
Maine Veterinary Medical Center
Medicine
Scarborough, ME, USA
Davis, CA, USA
Susan P. Gregory BVetMed PhD DVR DSAS (Soft Tissue)
Jacqueline Davidson DVM MS FHEA MRCVS
Clinical Professor Professor of Veterinary Nursing (retired)
Veterinary Medical Teaching Hospital, Texas A and M Royal Veterinary College, Department of Clinical Science
College Station, TX, USA and Services
North Mymms, Hatfield, Hertfordshire, UK
Daniel Degner DVM DACVS
Animal Surgical Center of Michigan Todd Hamilton VMD DACVS
Burton, MI, USA Maine Veterinary Medical Center
Scarborough, ME, USA
Ivan Doran BVSc CertSAS DSAS(Soft Tissue) MRCVS
RCVS Specialist in Small Animal Soft Tissue Surgery David Holt BVSc DACVS
Highcroft Veterinary Referrals Professor of Small Animal Surgery
Bristol, UK University of Pennsylvania, School of Veterinary Medicine
Philadelphia, PA, USA
Kenneth Drobatz DVM DACVECC
Professor of Small Animal Emergency and Critical Care Jennifer L. Huck DVM DACVS
University of Pennsylvania, School of Veterinary Assistant Professor of Surgery
Medicine University of Pennsylvania, School of Veterinary Medicine
Philadelphia, PA, USA Philadelphia, PA, USA
xiv Contributors

Georga T. Karbe Dr Med Vet MRCVS DACVS Mike McCallum CVT


Department Head: Soft Tissue Surgery Director of Operations
University of Veterinary Medicine Hannover, University of Pennsylvania, School of Veterinary Medicine
Hannover, Germany Philadelphia, PA, USA

Aarti Kathrani BVetMed PhD DACVIM DACVN FHEA MRCVS Meredith Maczuzak DVM DACVECC
Senior Lecturer in Small Animal Internal Medicine Emergency Medicine Specialist
Royal Veterinary College, Clinical Science and Services Pittsburgh Veterinary Specialty and Emergency
Department Center – BluePearl
Hatfield, Hertfordshire, UK South Hills, PA, USA

Deborah C. Mandell VMD DACVECC


Kristina Kiefer DVM PhD CCRP DACVSMR
Professor, Clinical Emergency and Critical Care
Surgical Coach
University of Pennsylvania, School of Veterinary Medicine
VetSSMART LLC
Philadelphia, PA, USA
St Paul, MN, USA
F.A. (Tony) Mann DVM MS DACVS DACVECC
Amie Koenig DVM, DACVIM DACVECC
Professor and Small Animal Soft Tissue Surgeon
Professor of Emergency and Critical Care
Director of Small Animal Emergency and Critical Care
Department of Small Animal Medicine and Surgery
Services
University of Georgia, College of Veterinary Medicine
University of Missouri-­Veterinary Medical Teaching
Athens, GA, USA
Hospital
Columbia, MO, USA
Amy Koenigshof DVM MS DACVECC
Two by Two Animal Hospital Anna Massie DVM DACVS
Berrien Springs, MI, USA Assistant Professor of Surgery
University of Pennsylvania, School of Veterinary Medicine
Janet J Kovak McClaren DVM DACVS DECVS MRCVS
Philadelphia, PA, USA
Staff Doctor
London Veterinary Specialists
Karol A. Mathews DVM DVSc DACVECC
London, UK
Professor Emeritus of Emergency and Critical Care
Ontario Veterinary College, University of Guelph
Elizabeth LaFond DVM DACVS DACVSMR
Guelph, ON, Canada
Veterinary Specialists of Sydney
Miranda, NSW, Australia Steve J. Mehler DVM DACVS
Main Line Veterinary Specialists
Nathaniel Lam DVM DACVS Devon, PA, USA
Chief of Surgery
VCA Oahu Veterinary Specialty Center Michael B. Mison DVM DACVS
Pearl City, HI, USA Surgery – MedVet Salt Lake City
Salt Lake City, UT, USA
Nyssa Levy DVM MS DACVECC
Assistant Professor, Emergency and Critical Care Medicine Eric Monnet DVM PhD FAHA DACVS ECVS
Department of Small Animal Clinical Sciences Professor of Surgery
Michigan State University, College of Veterinary Medicine Department of Clinical Sciences
East Lansing, MI, USA College of Veterinary Medicine and Clinical Sciences
Colorado State University
Lori Ludwig VMD MS DACVS Fort Collins, CO, USA
Mobile Veterinary Surgery
Charleston, SC, USA Pieter Nelissen DVM CertSAS DECVS MRCVS
European Specialist in Small Animal Surgery, RCVS
Cassie N. Lux DVM DACVS Recognized Specialist in Small Animal Surgery (Soft Tissue),
Associate Professor of Surgery Founder and Director, Department Head of Surgery
Department of Small Animal Clinical Sciences Frontier Veterinary Specialists
College of Veterinary Medicine, University of Tennessee Munich, Germany
Knoxville, TN, USA
Contributors xv

Matthew D. O’Donnell DVM DACVS Katrine Saile DVM, MS, DACVS


Central Texas Veterinary Specialty and Emergency Associate Surgeon
Hospital Blue Pearl Pittsburgh Veterinary Specialty and
Austin, TX, USA Emergency Center
Pittsburgh, PA, USA
Augusta Pelosi DVM DACVS DACVIM (Cardiology)
Cardiologist Jennifer Savini DVM DACVECC
Veterinary Heart Institute Emergency and Critical Care Specialist
Boca Raton, FL, USA VRC Specialty Hospital
Malvern, PA, USA
James A. Perry DVM PhD DACVIM (Oncology) DACVS
Veterinary Cancer and Surgery Specialists Mike Schlicksup DVM DACVS
Milwaukie, OR, USA Columbia Veterinary Emergency Trauma and Specialty
Columbia, SC, USA
Heidi Phillips VMD DACVS
Associate Professor of Small Animal Surgery Chad Schmeidt DVM DACVS
University of Illinois, College of Veterinary Medicine Professor of Small Animal Surgery
Urbana, IL, USA University of Georgia, College of Veterinary Medicine
Athens, GA, USA
David A. Puerto DVM DACVS
Center for Animal Referral and Emergency Services Deborah Silverstein DVM DACVECC
Langhorne, PA, USA Professor of Critical Care
University of Pennsylvania, School of Veterinary Medicine
MaryAnn Radlinsky DVM MS DACVS Philadelphia, PA, USA
Founding Fellow, Minimally Invasive Surgery
Small Animal Soft Tissue Ameet Singh BSc DVM DVSc DACVS
Salt River Veterinary Specialists Associate Professor of Surgery
Scottsdale, AZ, USA Ontario Veterinary College, University of Guelph
Guelph, ON, Canada
Erica Reineke VMD DACVECC
Associate Professor of Emergency and Critical Care Sean Smarick DVM DACVECC
University of Pennsylvania, School of Veterinary Independent Consultant
Medicine North Huntingdon, PA, USA
Philadelphia, PA, USA
Daniel D. Smeak DVM DACVS
Alexander M. Reiter Dipl Tzt Dr med vet DAVDC DEVDC Vice President, Surgical Models, Inotiv; Fort Collins
FF-­AVDC-­OMFS Emeritus Professor of Surgery
Professor of Dentistry and Oral Surgery Colorado State University
University of Pennsylvania, School of Veterinary Medicine Fort Collins, CO, USA
Philadelphia, PA, USA
Bryden J. Stanley BSc BVMS MACVSc MVetSc DACVS
Caryn Reynolds DVM DACVIM (Cardiology) Emeritus Professor
Cardiologist Michigan State University, College of Veterinary Medicine
Dove Lewis Emergency Hospital East Lansing, MI, USA
Portland, OR, USA
Vincent Thawley VMD, DACVECC
Jacob Rubin DVM DACVS Assistant Professor, Clinical Emergency and Critical Care
Maine Veterinary Medical Center University of Pennsylvania, School of Veterinary Medicine
Scarborough, ME, USA Philadelphia, PA, USA
xvi Contributors

Michael S. Tivers BVSc PhD CertSAS DECVS MRCVS Lori S. Wadell DVM DACVECC
Head of Surgery Professor of Clinical Critical Care Medicine
Paragon Veterinary Referrals University of Pennsylvania, School of Veterinary Medicine
Wakefield, UK Philadelphia, PA, USA

Karen M. Tobias DVM MS DACVS Mandy L. Wallace DVM MS DACVS


Institute Professor, Department of Small Animal Assistant Professor
Clinical Science University of Georgia, College of Veterinary Medicine
University of Tennessee, College of Veterinary Medicine Athens, GA, USA
Knoxville, TN, USA
Richard A.S. White BVetMed PhD DSAS DVR FRCVS
DACVS ECVS
Adam Townsend DVM
European and RCVS Specialist in Small Animal Surgery,
Resident, Small Animal Surgery
RCVS Specialist in Veterinary Oncology, ACVS Founding
University of Wisconsin-­Madison, School of Veterinary
Fellow, Surgical Oncology
Medicine
Clinical Director, Smart Paws GmbH
Madison, WI, USA
Special Professor of Small Animal Surgery
University of Nottingham, Nottingham, UK
Susan W. Volk VMD PhD DACVS
Associate Professor, Small Animal Surgery Chloe Wormser VMD DACVS
University of Pennsylvania, School of Veterinary Elite Veterinary Surgery
Medicine Northway Animal Emergency Clinic
Philadelphia, PA, USA Gansevoort, NY, USA
xvii

Preface

As the management of small animal surgical emergencies information on patient positioning and preparation in the
is constantly evolving, the purpose for writing this second operating room, essential equipment, and appropriate
edition is to have an up-­to-­date resource for the veterinary sterilization techniques. For the section on the
surgeon, critical care specialist, and general practitioner gastrointestinal system, four new chapters have been
that builds on information presented in the first edition added, including Rectal Prolapse, Colonic Torsion, Surgical
and provides updated information on preoperative Intervention Post-­Celiotomy, and a chapter on Enteral
stabilization, new and innovative treatment options and Feeding Tubes. Including the chapter on nutritional sup-
aftercare. Similar to the first edition, the majority of port reiterates the importance of appropriate stabilization
chapters are authored by two, and occasionally three, and aftercare to the ultimate success of the emergency sur-
specialists in the fields of both surgery and emergency/ gical patient. In the cardiovascular system section, a chap-
critical care to combine the expertise of individuals from ter has been added on Open-­Chest Cardiopulmonary
these two disciplines. In addition to approximately 70 Resuscitation, and in the section on wound management,
authors who contributed to the first edition, 18 new authors two new chapters have been added, including The Failed
have agreed to share their knowledge and expertise with Flap and Incisional Infections. Finally, for the section on
the reader. The layout of the second edition is similar to the orthopedic emergencies, in addition to updating chapters
first edition, with many of the chapters covering step-­by-­ from the first edition on the Approach to the Septic Joint
step information on emergency stabilization, diagnostic and the Surgical Management of Open Fractures, a chapter
approach, operative techniques, postoperative care, on Traumatic Joint luxation and Reduction has been added
common complications encountered, and how to to provide a comprehensive resource when faced with an
troubleshoot such complications should they occur. orthopedic emergency.
Relevant anatomy, imaging and full color illustrations and Although there are veterinary textbooks that focus on
drawings have been incorporated into each chapter to small animal surgery and those that focus on critical care
support the text. Case presentations have also been medicine, this is the only book that focuses on the
included when appropriate. combination of these two disciplines, filling a niche
To strive to enhance the content of the first edition, nine currently vacant in the veterinary literature. Additionally,
new chapters have been added based on advice received this book is unique in that the majority of the chapters are
from respected colleagues. Following the introductory co-­authored by experts in both fields. Because surgical
chapter, Triage and Initial Stabilization of the Emergency emergencies are common in veterinary medicine, this
Small Animal Surgical Patient, a new chapter, Operating textbook should have a wide audience, including surgical
Room Nursing Tips for Emergency Surgical Procedures, has and critical care residents in training, veterinary surgeons
been added to the beginning of the book to assist veterinary and criticalists, and veterinary practitioners who work in
nurses and operating room technicians who may be emergency medicine or those who have an interest in
working with these critical patients. This chapter provides surgery.
xviii

­Acknowledgements

I would like to thank the staff at Wiley, with special thanks to Meryl Le Roux and Erica Judisch, for all their help and
encouragement throughout this process. Without their support and guidance, completion of this project would not have
been possible.
The impact of Covid-­19 on many of the authors who contributed to this textbook cannot be overstated. The disruption in
normal work, family routine, health issues, and economic hardship resulted in significant fear and anxiety. I truly want to
thank all the authors for their dedication and commitment to this project – I know it wasn’t easy.
Finally, since the publication of the first edition, many of us lost a dear friend and colleague, Dr. Lesley King. Lesley was
a Professor of Critical Care at the University of Pennsylvania and was instrumental in the development of intensive care as
a veterinary specialty. Lesley made significant contributions to the veterinary literature including as a contributor to the
first edition of this textbook. Lesley was not only a colleague, but also a good friend, and will be dearly missed.
xix

­About the Companion Website

This book is accompanied by a companion website:

www.wiley.com/go/aronson/surgical

The website includes:


●● Case Studies
●● Video clips
●● References
xx

Daisy’s Story

ICU recovering. During times of COVID-­19, a new normal


developed and owners were unable to visit their pets dur-
ing their hospital stay. Daisy’s owners decided on the next
best thing – Facetime! Here she is being read a story during
one of her virtual visits with Mom and Dad.

Daisy’s story can be found in Chapter 54 (Case


Report 54.2). After an encounter with a porcupine, Daisy
underwent two major surgeries and spent weeks in our
1

Triage and Initial Stabilization of the Emergency Small Animal Surgical Patient
Dana Clarke
University of Pennsylvania, School of Veterinary Medicine, Philadephia, PA, USA

­Introduction which when combined with education of the medical staff


on the system’s guidelines for prioritizing medical care,
One of the most challenging aspects of emergency medi- reduces inconsistencies in decision-­making [3]. In veteri-
cine is being presented with patients who have a variety of nary medicine, no uniformly accepted triage system exists.
clinical signs and disease severities to assess, prioritize, sta- Veterinary healthcare professionals therefore use historical
bilize, and provide with definitive care. The veterinary cli- information and intuition to make rapid decisions regarding
nician is reliant on information provided by the client, the need for immediate care and order in which patients will
their physical assessment, and initial diagnostics to deter- be seen. The animal trauma triage (ATT) score was devel-
mine severity of illness and injury, and therefore, urgency oped retrospectively and assessed prospectively to help to
of care. Efficient identification and treatment of respira- classify and prognosticate for a heterogenous patient group.
tory, cardiovascular, urinary, and neurologic derangements For each patient, six categories are assessed (perfusion, car-
is essential for successful patient outcomes. When emer- diac, respiratory, skeletal, neurologic, and eye/muscle/integ-
gency surgical intervention is required, it is crucial to ument) and scored from 0 to 3, with 0 being unaffected or
appropriately stabilize the patient for anesthesia without only slightly affected to 3 indicating severe injury. The six
unduly delaying surgical care. scores are added together with a maximum possible score of
18. In both the retrospective and prospective populations,
the mean ATT score of survivors was significantly lower
­Triage and Initial Assessment than non-­survivors and for each one-­point increase in
ATT, the likelihood of survival decreased 2.3–2.6 times
Triage is an essential tool in the setting of emergency [4]. Another veterinary triage system, adapted from the
medicine to assess and prioritize critically ill patients [1, Manchester triage system, uses a five-­category system using
2]. This is particularly true of patients that may require color-­coding to indicate urgency. Examples of “red” emer-
emergency surgical intervention, as the time to provide gencies (those which need to be seen immediately) include
appropriate stabilizing care and definitive surgical ther- severe respiratory distress, decompensated shock, life-­
apy likely impacts patient outcome. threatening hemorrhage, and active seizures. Very urgent
In many veterinary hospitals, nurses obtain pertinent emergencies, including moderate respiratory distress, evi-
historical information and perform a basic assessment to dence of aortic thromboembolism, and urethral obstruction,
determine whether the patient needs immediate further were classified as “orange.” Urgent emergencies, such as
evaluation or is stable enough to be seen in turn. In general, mild hemorrhage, moderate dehydration and open fracture
over-­triage is preferred to under-­triage in veterinary medi- were classified as “yellow,” while non-­urgent disease pro-
cine, as the severity of signs presented by the patient and cesses such as localized inflammation, soft-­tissue swelling,
observed by the pet owner may not be fully appreciated by stranguria and recent isolated seizure were classified as
untrained individuals. Triage and training systems in patient “green” [5]. The study determined that the use of a veteri-
assessment are used routinely in human emergency medi- nary triage list by nurses upon triage corresponded better to
cine. A variety of triage systems exist for human patients, retrospectively reviewed patient status than when individual

Small Animal Surgical Emergencies, Second Edition. Edited by Lillian R. Aronson.


© 2022 John Wiley & Sons, Inc. Published 2022 by John Wiley & Sons, Inc.
Companion website: www.wiley.com/go/aronson/surgical
2 Triage and Initial Stabilization of the Emergency Small Animal Surgical Patient

judgment and intuition upon triage were employed [5].


Multicenter, prospective evaluation of this veterinary triage
list is warranted to determine whether patient care is
improved and time to be seen can be better estimated.
Irrespective of the need for a formal veterinary triage sys-
tem, a brief, but thorough physical exam remains the gold
standard for recognizing critical patient status. The initial
triage assessment includes visual examination and assess-
ment of four key body systems: cardiovascular, respiratory,
neurologic, and urinary [6, 7]. Information regarding the
patient’s mentation and responsiveness, as well as respira-
tory rate and effort, are obtained quickly on brief visual
exam, often before performing any parts of a physical
examination. Thoracic and cardiac auscultation with con-
current pulse palpation and a more thorough assessment
of neurologic status, if indicated, follows visual examina- Figure 1.1 Cat with open mouth breathing secondary to the
pain associated with an aortic thromboembolism.
tion. After cardiovascular, respiratory, and neurologic sta-
tus is determined, if the patient is stable enough for further
evaluation, urinary triage can be performed. Any signifi- foreign body should be ruled out. See Chapter 28 for stabi-
cant pain must be addressed urgently to improve patient lization of the patient with upper airway obstruction.
comfort and so that the effects of pain do not alter interpre- If decreased lung sounds relative to respiratory effort are
tation of cardiovascular and respiratory findings. heard dorsally, pneumothorax should be suspected.
Additionally, aggression should not be considered a sign of However, if decreased lung sounds are heard ventrally,
patient stability, as many scared and stressed patients will pleural effusion should be considered. Lung sounds may
be aggressive in the face of severe shock. not be completely absent in the presence of pleural effu-
sion or pneumothorax, they may be reduced relative to the
other lung fields but still present. See Chapter 34 for stabi-
­Respiratory Assessment lization of the patient with pleural space disease. Diseases
of the chest wall such as masses, rib fractures, and flail seg-
Before any physical examination, all patients should have ments may also result in abnormal auscultation of the pul-
their respiratory rate, effort, noise, and pattern observed monary parenchyma. For more information on chest wall
from afar. If a patient is showing any changes in respira- disease, see Chapter 41.
tory pattern or effort, oxygen supplementation should be For patients in respiratory distress with a heart murmur,
provided immediately. If there is any respiratory compro- arrhythmia, pleural effusion, or pulmonary crackles pre-
mise, the patient should be presumed to be in hypoxemic sent on auscultation, cardiogenic and hypoxemic shock
shock until proven otherwise and oxygen supplementa- should be considered as possible differentials. This is par-
tion should be provided. Further respiratory triage involves ticularly important since fluid therapy is often contraindi-
auscultation of the upper airway, trachea, and thorax. cated in most patients with cardiac dysfunction or failure
During abbreviated thoracic auscultation, emphasis and must be ruled out, to the best of the clinician’s ability
should be placed on determining heart rate, rhythm, the on triage, prior to administering intravenous (IV) fluid
presence of murmur(s) or arrhythmia(s), and lung sounds therapy.
in all lung fields. Visual and auditory assessment of res-
piratory pattern and noise combined with thoracic auscul-
Hypoxemic Shock
tation should help localize the anatomic origin of the
respiratory distress. The impact of pain, stress, and anxiety Hypoxemic shock occurs secondary to decreased arterial
on respiratory rate and effort should not be underesti- blood oxygen content. Common causes of hypoxemic shock
mated (Figure 1.1). include pulmonary parenchymal disease, such as pneumo-
If respiratory noise is localized to the upper airway, dis- nia, severe anemia, and hypoventilation (Figure 1.2). Many
eases associated with an upper airway obstruction, includ- veterinary patients in hypoxemic shock are at the limits of
ing laryngeal paralysis, laryngeal collapse, brachycephalic their physiologic reserves, and are intolerant of excessive
airway disease, tracheal collapse, the presence of a tra- handling, restraint, and manipulation; they should be han-
cheal, laryngeal, or pharyngeal mass, and the presence of a dled carefully. Clinical signs include weakness, mental
­Respiratory Assessmen  3

Figure 1.2 Lateral thoracic radiograph showing cranioventral


pulmonary infiltrates creating an alveolar pattern consistent
with aspiration pneumonia.

depression, pale mucous membranes, dyspnea, crackles or


increased bronchovesicular lung sounds, or decreased lung
sounds ventrally (pleural effusion) or dorsally (pneumotho-
rax), and cyanosis. Patients with diaphragmatic hernia may
have decreased lung sounds dorsally or ventrally. Cyanosis
Figure 1.3 Continuous pulse oximetry assessment in a laterally
is only seen with severe hypoxemia (at least 5 g/dl of deoxy-
recumbent dog receiving oxygen supplementation via
genated hemoglobin), and thus the absence of cyanosis nasal prongs.
absolutely does not rule out hypoxemia. In anemic animals,
cyanosis is unlikely to be detected due to decreased hemo- (PaO2) consistent with hypoxemia, and an increased par-
globin concentration, and therefore should not be relied tial pressure of alveolar–arterial oxygen gradient P(A–a)
upon to diagnose hypoxemia [8]. O2). Calculation of the P(A–a)O2 gradient provides objec-
In any patient with suspected hypoxemic shock, sup- tive information on pulmonary function by removing the
plemental oxygen should be provided until the ability to influence of ventilation on PaO2. When a patient is
adequately oxygenate is confirmed. Diagnostics that can breathing 21% oxygen, the P(A–­a)O2 should be less than
be helpful for the patient in hypoxemic shock include 10–15 mmHg. When a patient is breathing 100% oxygen,
pulse oximetry (peripheral capillary oxygen saturation, the P(A–a)O2 should be less than 150 mmHg. If the
SpO2), arterial blood gas analysis, thoracic radiographs, P(A–a)O2 gradient is greater than 15 mmHg while breath-
thoracic/trauma computed tomography (CT), and tho- ing 21% oxygen, it is consistent with pulmonary dysfunc-
racic ultrasound. SpO2 may be less effective with bright tion. For A–a gradient calculation, see the formula
lighting, poor perfusion, high motion, and pigmentation in Box 1.1.
of the skin. It is convenient since it can noninvasively Preliminary evaluation of the ratio of SpO2 to fraction of
determine percentage of oxygenated hemoglobin, and, inspired oxygen (FiO2) to the partial pressure of oxygen in
for sedentary patients, can be left in place for continuous arterial blood to FiO2 (PaO2/FiO2) showed good correlation
monitoring (Figure 1.3). Many patients in respiratory dis- between the two values in dogs. It is possible that with fur-
tress will not tolerate the restraint necessary for arterial ther investigation, the SpO2/FiO2 may become a reliable,
blood gas collection and thoracic radiographs, especially less invasive alternative to determining PaO2/FiO2 [9].
on presentation. If obtaining an arterial blood gas is fea- Thoracic radiographs may show pulmonary parenchy-
sible, findings may include decreased SpO2, decreased mal infiltrates ventrally consistent with pneumonia,
partial pressure of carbon dioxide (PaCO2) consistent caudodorsally consistent with non-­cardiogenic pulmo-
with hyperventilation, increased PaCO2 consistent with nary edema, and in the perihilar region consistent with
hypoventilation, decreased partial pressure of oxygen congestive heart failure. In the trauma patient,
4 Triage and Initial Stabilization of the Emergency Small Animal Surgical Patient

and pericardial effusion, pneumothorax, and pulmonary


Box 1.1 Formula Used for A–a Gradient Calculation
parenchymal infiltrates [15–20]. It is particularly useful
The partial pressure of alveolar–arterial oxygen in patients that are not stable enough for thoracic radio-
(P(A–a)O2) gradient provides objective information on graphs, as well as a monitoring tool to assess for response
pulmonary function by removing the influence of venti- to therapy. Thoracic ultrasound may be performed with
lation on PaO2. the patient in sternal or lateral recumbency. Pleural effu-
P A-a O2 PAO2 PaO2 sion is generally visible in the cranial and/or caudoven-
tral pleural space. Ultrasound guidance to localized fluid
PAO2 FO2
i Patm PH2O PaCO2 / R
pockets can be helpful to guide thoracocentesis. When
PAO2 = alveolar gas equation evaluating for the presence of pneumothorax, the caudo-
FiO2 = percentage of inspired O2 dorsal thorax is evaluated for the lack of a “glide” sign,
Patm = atmospheric pressure (760 mmHg used at which is diagnostic for pneumothorax. A glide sign is
sea level) created by the normal back and forth respiratory motion
PH2O = water vapor pressure (53 mmHg 39°C for dogs/ of the interface between the visceral and parietal pleura
cats; 47 mmHg at 37°C in humans) (Video 1.1). Free air in the thoracic cavity obliterates the
R = respiratory quotient (approximately 0.8–0.9) glide sign [15–17]. Cellular or fluid infiltrate into the
pulmonary parenchyma, as with edema, hemorrhage,
Example Blood Gas
and pneumonia can be assessed using ultrasound in four
PaCO2 = 24.2 windows in each hemithorax (caudodorsal, cranial, mid-
PaO2 = 59.5 dle lung lobe regions, and perihilar) for the presence of
PAO2 = (0.21 × (760 – 53)) – (24.2/0.9) increased penetration of ultrasound, which manifest as
PAO2 = 121.6 hyperechoic lines (B-­lines) in parallel with the ultra-
P(A–a)O2 = 121.6–59.5 sound beam, that can be individual or coalescing
P(A–a)O2 = 62.1 (indicates hypoxemia is due to (Figure 1.4 and Video 1.2) [18–22].
pu­lmonary dysfunction)
Video 1.1 TFAST showing a normal glide sign, which is
created by the respiratory motion of the visceral and pari-
etal pleural interface sliding back and forth.
pulmonary contusions, which can be present in any lung
field(s), may not become radiographically apparent for
Video 1.2 TFAST showing coalescing B-­lines created by
up to 48 hours, although peak opacification has been
marked pulmonary infiltrates allowing ultrasound pene-
shown to occur at 6 hours in human trauma patients [10].
tration into the pulmonary parenchyma.
Additionally, up to 30% of human trauma patients do not
have radiographic evidence of contusions on initial tho-
racic radiographs, which is why CT is often proposed as
the preferred method of thoracic imaging [10–12]. In a
study of dogs that had succumb to vehicular trauma,
thoracic radiographs underestimated the presence of
contusions, while also overestimating their severity. The
same study also noted that thoracic radiographs were
less sensitive than CT for detecting rib fractures [13].
Initial investigation in the use of thoracic ultrasound for
detection of pulmonary contusions in dogs with vehicu-
lar trauma showed a high sensitivity for diagnosing con-
tusions compared with CT, and even noted improved
sensitivity compared with thoracic radiographs [14].
Therefore, cautious respiratory monitoring and repeat
thoracic imaging may be indicated in any patient with a
history of known or suspected trauma.
Thoracic ultrasound, also known as thoracic focused
Figure 1.4 TFAST ultrasonographic appearance (still image) of
assessment with sonography for trauma, triage, and a B-­line, which is created by increased infiltrates in the
tracking (TFAST), allows clinicians to assess for pleural pulmonary parenchyma allowing ultrasound penetration.
­Cardiovascular Assessmen  5

­Cardiovascular Assessment direct measurements [28]. Direct arterial blood pressure


is considered the gold standard for blood pressure deter-
The most important part of the cardiovascular assessment mination, and offers the additional benefits of continu-
during emergency patient triage is the determination ous, real-­time results that are accurate with arrhythmias
whether the patient is in shock. If shock is suspected, the and decreased perfusion. However, placement of an arte-
type of shock and need for fluid therapy must then be rial catheter is technically challenging, especially in dis-
determined. The common feature in all shock patients is tressed or hypotensive patients and cats, uncomfortable
inadequate cellular energy metabolism, which is most for the patient during placement, and requires constant
commonly due to poor perfusion. However, metabolic and monitoring to ensure the catheter is not inadvertently
hypoxic shock can occur with normal perfusion, so one dislodged. ECG is helpful to evaluate for the presence of
must be careful to not rule out shock on the basis of normal cardiac arrhythmias, which can be the primary cause of
perfusion parameters alone [23, 24]. shock (cardiogenic), or secondary complications of hypo-
For cardiovascular triage, mucous membrane color, tem- volemic, metabolic, hypoxic, or distributive shock.
perature, and capillary refill time (CRT) can be used to Venous blood gas monitoring, particularly for pH, partial
assess perfusion. Signs of poor perfusion during mucous pressure of carbon dioxide in venous blood (PvCO2), par-
membrane assessment include pale pink to white mucous tial pressure of oxygen in venous blood (PvO2), electro-
membranes, cool temperature, and prolonged to absent lytes, and base excess/deficit is important to help
CRT (> 2 seconds). Bright pink or red mucous membranes, determine the underlying cause of cardiovascular com-
injected capillaries, and rapid CRT can be seen with dis- promise, assess cellular oxygen delivery and metabolism,
tributive shock. However, depending on the patient’s stage and response to therapy. Similarly, PCV/TS are essential
of cardiovascular compromise and the degree of compen- to evaluate for blood and/or protein loss, dehydration,
sation, even patients with shock can have normal mucous and appropriate hemodilution response if fluid therapy
membrane appearance. Heart rate and rhythm should be is used. Lactate can be a marker of anaerobic metabolism
assessed simultaneously with pulse palpation to determine and is often increased in shock patients (type A lactic aci-
pulse pressure quality and for deficits. Both femoral and dosis), although less reliably in cats. It can support clini-
dorsal metatarsal artery palpation is preferred to appreciate cal assessment of poor perfusion and trended over time
discrepancies in proximal and distal perfusion. Extremity with treatment of the primary cardiovascular distur-
temperature on limb palpation and rectal temperature bance. It has been associated with outcome in gastric
should be noted to complete the patient’s perfusion clinical dilatation and volvulus, pyometra, and immune-­
picture [23–26]. mediated hemolytic anemia [29–35]. It is important to
Following physical examination of the cardiovascular remember that type B lactic acidosis, which is hyperlac-
system, emergency diagnostic tools that can aid cardio- tatemia in the face of normal perfusion, does not resolve
vascular triage include indirect blood pressure, electro- with fluid therapy. Causes of type B lactic acidosis
cardiogram (ECG), venous or arterial blood gas, packed include liver failure, neoplasia (especially hematopoi-
cell volume/total solids (PCV/TS), lactate, and left atrial etic), diabetes mellitus, sepsis/systemic inflammatory
to aortic root ratio on ultrasound. Indirect blood pressure response syndrome, and various drugs and toxins [30].
methods, such as Doppler or oscillometric technologies, Comparing the left atrium diameter with the root of the
provide rapid noninvasive determination of arterial aorta (LA : Ao), when using a short axis view from the right
blood pressure. Doppler is particularly useful in small parasternum, is helpful to assess left atrial volume. The
patients, cats, and those with cardiac arrhythmias. LA : Ao ratio was originally developed to support a diagnosis
Oscillometric methods are convenient as they can be pro- of congestive heart failure, but it can also be used to evaluate
grammed to cycle at predetermined intervals, such that for left atrial volume underload, which can occur with hypo-
repeated measurements can be obtained automatically. volemic shock. In dogs, the normal LA : Ao ratio is 1.3,
For both methods, cuff size selection in relation to limb whereas the ratio is 1.5 in cats [36, 37]. Baseline LA:Ao ratio
diameter is essential for accurate results. Cuff diameter on triage can help support a diagnosis of cardiogenic shock
should be approximately 40% of the limb circumference secondary to congestive heart failure if the LA : Ao ratio is
in dogs and 30% in cats. Cuffs that are too large will gen- increased. When the ratio is decreased, hypovolemic shock
erate falsely low blood pressure results, and falsely high may be present, especially if found in conjunction with other
results will be obtained from a cuff that is too small [27]. parameters that support hypoperfusion. Changes in the ratio
In hypotensive patients, noninvasive methods have been with treatment, whether intravenous fluids or diuretics, can
shown to have the greatest variability compared with be useful for monitoring response to therapy.
6 Triage and Initial Stabilization of the Emergency Small Animal Surgical Patient

The use of point of care ultrasound techniques in dogs as hetastarch, due to concerns for increased mortality,
has been used to objectively assess vascular status and severe renal injury, and risk of bleeding associated with
may be used as an adjunct diagnostic to guide fluid and/or their use in critically ill adults, including those with sepsis
vasopressor therapy. In normal dogs with furosemide and admitted to the intensive care unit. VetStarch® (Abbott
induced hypovolemia, the caudal vena cava to aorta ratio Laboratories, Chicago, IL), a veterinary specific HES solu-
measured from the right intercostal space in left lateral tion, is commercially available as a synthetic colloid for
recumbency corresponded with decreasing body weight plasma volume expansion. Preliminary veterinary studies
consistent with volume loss [38]. This ratio was also have conflicting evidence on association between the use
decreased after blood donation in healthy dogs [39]. In of synthetic colloids and acute kidney injury in dogs; they
greyhounds, the iliac location for this ratio did not detect should be used with caution until further research is avail-
volume status changes before and after blood dona- able [48, 49].
tion [40]. While additional work is needed in canine clini- Hypovolemic resuscitation or controlled intravascular
cal cases of hypovolemia as well as normal and critically ill volume replacement titrated to a mean arterial blood pres-
cats, this technique appears to be a promising method for sure (MAP) of 60 mmHg is widely used for human and vet-
non-­invasive volume assessment. erinary patients with hemorrhagic shock [50–54]. The goal
is to preserve perfusion to the vital organs, particularly the
kidneys and cerebral circulation, without supranormaliz-
Hypovolemic Shock and Fluid Therapy
ing blood pressure, to prevent disruption of any clots tem-
Hypovolemic shock, which is the most common type of shock pering further hemorrhage. Experimental evidence in a
seen in veterinary medicine, can be due to blood loss, fluid swine model shows that rebleeding occurs when MAP is
loss, or inadequate intake, and results in decreased tissue greater than 60 mmHg, while maintaining the MAP at
delivery of oxygen. Clinical signs consistent with hypovolemic approximately 60 mmHg maintains renal and cerebral
shock are mental depression, weakness, pale mucous mem- blood flow [54]. Recommendations for decreased volume
branes, prolonged CRT, tachycardia, weak peripheral pulses, fluid resuscitation for crystalloid boluses are between 20
cool extremities, and tachypnea. and 30 ml/kg and 5 ml/kg for colloid boluses titrated to
To treat hypovolemic shock, intravascular volume effect and target blood pressure [52].
replacement is essential and generally accomplished with Transfusion with packed red blood cells (pRBC; 5–10 ml/
intravenous crystalloids, colloids, blood products, or a kg), fresh frozen plasma (FFP; 10–20 ml/kg), or whole
combination of the fluid replacement options. “Shock” blood (10–20 ml/kg) may be indicated for patients with
doses of fluid therapy are based on the blood volume for a anemia and/or coagulopathy. While there is no absolute
given species, and amounts for replacement are based on PCV below which a transfusion is required, consideration
the percentages of volume loss to cause cardiovascular of the chronicity of anemia, cardiovascular stability, con-
changes secondary to hypovolemic shock. Blood volume is tinuing losses, anticipated surgical intervention, and pul-
approximately 90 ml/kg in dogs and 45–60 ml/kg in cats. monary function all impact the decision of whether or not
Generally, patients are given portions of their shock dose to transfuse a patient. It is also important to remember, that
of fluids, such as 10–30 ml/kg of balanced isotonic crystal- in many critically ill patients, even after control of hemor-
loid solutions or 5–10 ml/kg of colloid solutions as a bolus rhage, coagulopathy may persist due to dilution, consump-
over 15–20 minutes and assessed for improvement in per- tion, delayed liver production of clotting factors, and liver
fusion parameters. The bolus is repeated if indicated. dysfunction, so repeated dosing of FFP may be needed
Hypertonic saline (7.5%, 3–5 ml/kg IV over 15–20 minutes even once coagulation parameters have normalized.
in dogs, 2–3 ml/kg IV over 15–20 minutes in cats) is also Regardless of the fluid type chosen for cardiovascular
effective for rapid volume expansion in hypovolemic resuscitation, it is imperative that frequent reassessment of
shock but should only be used in patients with normal the patient’s cardiovascular parameters in response to treat-
hydration. The decision about whether crystalloids or col- ment be performed. That same physical exam parameters
loids should be chosen as the initial resuscitation fluid is and initial diagnostics used to diagnose shock should be
controversial and has yet to be determined in both human reevaluated. Additional diagnostics that may be helpful for
and veterinary medicine [41–47]. In veterinary patients, determining whether a patient is appropriately or maximally
the decision is often based on availability, cost, and fluid resuscitated, especially if shock persists, include cen-
whether there are concerns about the patient’s colloid tral venous pressure (CVP) and central venous oxygen satu-
osmotic pressure and the ability to maintain fluid within ration (SCVO2). CVP, which is a measure of the hydrostatic
the intravascular space. In June 2013, a boxed warning pressure within the intrathoracic (cranial or caudal) vena
was placed on hydroxyethyl starch (HES) solutions, such cava, is used to approximate right atrial pressure, or preload.
­Cardiovascular Assessmen  7

Normal CVP is 0–5 cm H2O. CVP values less than 0 cm H2O respiratory rate or effort, respiratory distress, pulmonary
are consistent with hypovolemia or decreased venous tone crackles (pulmonary edema), and decreased lung sounds
secondary to vasodilation [55]. Increased CVP (> 7–10 cm ventrally consistent with pleural effusion (cats).
H2O) can be seen with volume overload, pleural space dis- In addition to history and physical examination find-
ease (pneumothorax, pleural effusion), pericardial disease ings, other diagnostics often needed to diagnose cardio-
(restrictive pericarditis, pericardial effusion), tricuspid valve genic shock include ECG, blood pressure, pulse oximetry
disease, myocardial disease, and intraabdominal hyperten- (SpO2), thoracic radiography, and TFAST. TFAST can be
sion [55, 56]. The value of CVP monitoring for guiding fluid used to determine cardiac contractility, myocardial
resuscitation has been questioned in both human and veteri- thickness, and cardiac chamber (atria and ventricle)
nary critical care in recent years. In addition to CVP, central size. Focused echocardiography training for emergency
catheters can also be used to measure SCVO2, which is an veterinarians has been shown to improve their diagnos-
assessment of global tissue oxygenation and is the percent- tic capabilities for determination of several cardiac
age of saturated hemoglobin within the cranial or caudal abnormalities [59]. Treatment may involve oxygen sup-
vena cava or right atrium. Alterations in SCVO2 reflects plementation, pericardiocentesis, diuretic therapy, anti-­
imbalance between oxygen delivery and consumption. arrhythmics, vasopressors, or vasodilators depending on
Decreased SCVO2 is seen with increased oxygen consump- the etiology of cardiogenic shock.
tion relative to delivery, as with hypovolemia, anemia, car-
diac dysfunction, pulmonary dysfunction, fever, and
Distributive/Septic Shock
hyperthermia. Increased SCVO2 is seen with decreased oxy-
gen consumption relative to delivery, as with hypothermia Distributive shock is defined as a maldistribution of blood
and mitochondrial dysfunction [57–59]. In critically ill dogs, flow, most commonly due to altered systemic vascular
a decrease in SCVO2 below 68% within the first 24 hours of resistance (SVR). Decreased SVR is the most common
hospitalization was associated with poor outcome with pro- SVR alteration, and vasodilatory shock secondary to sep-
gressive increase in mortality with decrease in SCVO2 [59]. In sis is one of the most readily recognized forms of distribu-
septic dogs that underwent surgery for pyometra, survivors tive shock. Distributive shock can also be secondary to
had lower lactate, base deficit and their average SCVO2 was obstructive disease processes, such as gastric dilation and
74.6%. Non-­survivors in this study had an average SCVO2 of volvulus, pericardial effusion, and neoplasia causing vas-
62.4% [33]. Co-­oximetry, which is not widely available, is cular obstruction (such as adrenal tumors with invasion
needed for SCVO2 determination; this is likely the reason for into the vena cava) [25]. The clinical signs of distributive
its limited clinical use in veterinary patients. shock in dogs are often very different from other forms of
shock. In dogs, the mucous membranes are often bright
pink (Figure 1.5), CRT is decreased (< 2 seconds), and
Cardiogenic Shock
peripheral pulses can be bounding or more prominent
It is important to differentiate hypovolemic shock from than normal. Cats with septic shock generally do not
cardiogenic shock, as many of the physical exam find- demonstrate the hyperdynamic signs seen in dogs and
ings can overlap but the treatment is usually vastly dif- instead have pale mucous membranes, bradycardia, and
ferent. Fluid therapy is generally contraindicated in decreased rectal temperature [60].
most patients with cardiogenic shock. Cardiogenic Many patients with distributive shock also have a com-
shock can be due to forward (left-­sided) or backward ponent of hypovolemic shock (absolute or relative), so
(right-­sided) failure of blood flow. Common causes of fluid therapy to correct intravascular volume deficit is
cardiogenic shock include congestive heart failure, essential. In humans with severe sepsis and septic shock,
s­ystolic dysfunction, as with dilated cardiomyopathy, early goal-­directed therapy is shown to improve patient
diastolic dysfunction, as with hypertrophic cardiomyo- outcome when compared to traditional management strat-
pathy, and arrhythmias [25, 26]. Clinical signs of cardio- egies. In two landmark human studies, hemodynamic
genic shock include pale mucous membranes, heart parameters such as direct arterial blood pressure, CVP, and
murmur and/or arrhythmias, poor or variable pulse SCVO2 measurement, and treatment with crystalloids, col-
quality, pulse deficits, and tachycardia or bradycardia. loids, pRBC, and catecholamines to improve cardiac con-
Findings consistent with right-­sided heart failure tractility and/or vasomotor tone were used until prescribed
include decreased ventral lung sounds consistent with endpoints were achieved [61, 62]. Standardized goal-­
pleural effusion, jugular venous distension, ascites, and directed therapy does not yet exist for veterinary patients,
hepatomegaly. Clinical signs seen with left-­sided dys- therefore, normalization of routinely monitored cardiovas-
function and left-­sided heart failure include increased cular and perfusion parameters, including heart rate and
8 Triage and Initial Stabilization of the Emergency Small Animal Surgical Patient

therapy, mortality decreased from 33.3% to 19.5% [69]. In


veterinary patients with septic shock, antimicrobials
should be given as soon as reasonably possible, especially
for those that will undergo emergency anesthesia and sur-
gery. In critically ill septic patients anticipated to undergo
surgery, perioperative first-­ and second-­generation cepha-
losporins likely do not provide adequate antimicrobial
coverage and should not be used in favor of more broad-­
spectrum medications. In the absence of confirmatory cul-
ture and sensitivity testing, broad-­spectrum therapy should
be used until a diagnostic culture result is obtained and
antimicrobial therapy can be de-­escalated. Intravenous
administration is preferred in all cardiovascularly unstable
and critically ill patients as oral, intramuscular, and subcu-
taneous absorption may not be predictable. Antibiotics that
can be considered for first-­line broad-­spectrum therapy
include ampicillin and clavulanate (Unasyn®, Pfizer,
22–30 mg/kg IV every 8 hours); ampicillin (18–22 mg/kg IV
every 8 hours) combined with enrofloxacin (10–15 mg/kg
IV every 24 hours), cefoxitin (30 mg/kg IV every 6 hours),
and clindamycin (10 mg/kg IV every 12 hours) combined
with cefotaxime (40–50 mg/kg IV every 6 hours), or ceftazi-
dime (30–50 mg/kg IV every 6–8 hours with dosing at the
lower end of the range for cats and higher end for dogs).

Figure 1.5 Bright pink mucous membranes in a dog with septic


peritonitis. Metabolic Shock
Metabolic shock is defined as dysfunction of cellular
rhythm, rectal temperature, mucous membrane color and metabolism, which generally occurs in the face of ade-
CRT, blood pressure, CVP, PCV/TS, lactate and base deficit quate perfusion and oxygenation. Examples include
are recommended [31, 63, 64]. severe pH derangements, hypoglycemia, adrenal insuf-
When fluid therapy fails to normalize hemodynamic ficiency, and certain toxicities such as cyanide. The clini-
parameters, particularly blood pressure in septic patients, cal signs seen with metabolic shock closely resemble
vasoactive catecholamines may be necessary [65, 66]. those in hypovolemic shock and are related to the under-
Commonly used vasoactive catecholamines used in critical lying etiology. Mental depression is the most universally
care are dopamine, dobutamine, and norepinephrine recognized sign of metabolic shock. The same diagnos-
(Table 1.1). Vasopressin, also known as antidiuretic hor- tics used for patients in hypovolemic shock are indicated
mone, is a peptide synthesized in the pituitary that binds for the patient in metabolic shock. Treatment may
vasopressin specific receptors on vascular smooth muscle. include correction of acid–base derangements with IV
Vasopressin stores can become depleted with prolonged fluids and/or bicarbonate, dextrose supplementation,
shock or sepsis resulting in vasoplegia despite intravenous and steroid administration, if adrenal dysfunction is
fluid and vasoactive catecholamine therapy. Vasopressin demonstrated or highly suspected [70–74].
deficiency has been documented in people with refractory
hypotension, and positive benefit has been shown with the
addition of intravenous administration of vasopressin. ­Dehydration
Experience with vasopressin is growing in veterinary medi-
cine [67, 68]. The management of hypovolemic shock focuses on resto-
Early administration of broad-­spectrum antibiotics has ration of intravascular volume for improvement in
been shown to improve survival in human patients with cardiovascular function, rather than normalization of
sepsis and septic shock when combined with early goal-­ hydration. Dehydration is a reflection of interstitial fluid
directed therapy. When antibiotics were given within one balance, and while imperative to assess, treat, and monitor
hour of triage in combination with early goal-­directed for improvement, treating hypovolemic shock must take
­Dehydratio  9

Table 1.1 Commonly used vasopressors used in the emergency room and intensive care unit.

CRI dose Effect Additional information

Dopamine 1–4 μg/kg/minute Vasodilation (renal) Mixed data for renal effects
5–10 μg/kg/minute Increased contractility, some
vasoconstriction
10–20 μg/kg/minute Vasoconstriction, variable contractility
effects
Dobutamine 2–20 μg/kg/minute (dogs) Increased contractility, little
vasoconstriction
2–5 μg/kg/minute (cats) Increased contractility, little Can cause seizures in cats
vasoconstriction
Norepinephrine 0.05–2 μg/kg/minute Potent vasoconstriction
Vasopressin 0.5–2 mU/kg/minute (dogs) Potent vasoconstriction (even in acidosis) Limited clinical experience in
dogs, no dose established for cats

Source: Adapted from Simmons and Wohl [65]. CRI = constant rate infusion.

priority. Interstitial fluid losses are generally gradual and especially cats, as significant overhydration can result if
are therefore corrected over time. Prolonged or severe overweight or obese body weight is used. The deficit is
dehydration can lead to hypovolemic shock. The percent- then corrected over a period of 12–48 hours depending on
age of dehydration (5–12%) is estimate based on physical chronicity, patient’s tolerance to fluid therapy, mainte-
exam (skin turgor, sunken eyes, urine output) and objec- nance fluid needs, and any continuing fluid losses
tive criteria, such as PCV/TS, loss of body weight, and (Box 1.2). In small animal patients, maintenance fluid
urine specific gravity. The fluid deficit is determined using rates are generally 2–3 ml/kg/hour in dogs and 1–2 ml/kg/
the percentage of dehydration and the patient’s lean body hour in cats [75].
weight. Estimation of lean body weight is imperative Electrolyte monitoring should be performed routinely
when determining the fluid prescription for obese patients, (Table 1.2) in patients with dehydration and shock. This is

Box 1.2 Fluid Therapy Prescription Formula


This formula incorporates fluid deficit (dehydration), ongoing losses, and maintenance fluid needs. It should be used
only after intravascular volume deficits (hypovolemic shock) have been corrected.
Fluid prescription fluid deficit liters continuing losses including insensible losses maintenance fluid needs
Fluid deficit liters %dehydration lean body weight kg
Rate of deficit correction is generally over 12–36 hours depending on patient stability, chronicity of dehydration, and
tolerance for IV fluids.
Maintenance needs are generally 2–3 ml/kg/hour for dogs and 1–2 ml/kg/hour for cats.
Example Fluid Prescription Calculation
25 kg mixed breed dog (lean body condition)
Estimated to be 8% dehydrated based on physical exam findings (tacky mucous membranes, prolonged skin tent,
slightly sunken globes, hyperviscous saliva in the corner of the mouth).
No conditions that would make the patient fluid intolerant; plan to correct over 24 hours.
The dog is losing approximately 60 ml in vomit every hour, no excessive gastrointestinal or urinary losses.
Deficit = 0.08 × 25
Deficit = 2000 ml
Rate of deficit correction = 2000/24 = 83 ml/hour
Fluid prescription (per hour) = 83 ml (deficit) + 60 ml (losses) + 50 ml (maintenance)
Fluid prescription = 193 ml/hour
Table 1.2 Monitoring parameter guidelines and frequencies for dehydrated patients and those in hypovolemic, distributive, and hypoxemic shock.

Urine output/
Physical assessmenta Blood pressure SpO2 specific gravity PCV/TS/BG/Azo Stick® VBG/ABG/electrolytes

Dehydrationb 8–12 hours 8–12 hours 12–24 hours 8–12 hours 12–24 hours 12–24 hours
Hypovolemic 1–2 hours initially, then 4–6 hours 1–2 hours initially, 4–6 hours 4–6 hours 4–6 hours initially, 4–6 hours initially, then
shock once stabilized then 4–6 hours then 6–8 hours 6–8 hours
Distributive 1–2 hours initially, then 4–6 hours 1–2 hours initially, 4–6 hours 4–6 hours 6–8 hours 6–8 hours
shock once stabilized then 4–6 hours
Hypoxemic shock 1–2 hours initially, then 4–6 hours 2–6 hoursc 1–4 hoursc 4–6 hoursc 12–24 hours and after 12–24 hoursc
once stabilizedc pRBC transfusionc
a
Physical assessment parameters include hydration evaluation, mucous membranes, capillary refill time, respiratory rate and effort, cardiac and thoracic auscultation, pulse quality, and
temperature.
b
Dehydrated patients should also be weighed every 8–12 hours.
c
Frequency of diagnostics will depend on patient stability and amount of stress caused to the patient with handling, evaluation, and blood sampling.
ABG, arterial blood gases; BG, blood glucose; PCV, packed cell volume; SpO2, peripheral capillary oxygen saturation; TS, total solids; VBG, venous blood gases.

0005276422.INDD 10 04-15-2022 07:54:22


Table 1.3 Analgesics, sedatives, and anxiolytics used in small animal medicine.

Generic drug Brand (manufacturer) Dose Comment

Opioids:
Buprenorphine Buprenex® (Reckitt & 5–20 μg/kg IM, IV q 6–8 hours μ-­partial agonist
Colman) Cats: 10–20 μg/kg PO q 6–8 hours Excellent oral absorption (cats)
Difficult to reverse
Butorphanol Torbutrol®, Torbugesic-­SA® 0.1–0.4 mg/kg IM, IV q 1-­4hours κ-­agonists
(Zoetis) Partial μ reversal: 0.05–0.1 mg/kg IV μ-­antagonist
CRI loading dose: 0.1 mg/kg IV Variable analgesia
CRI: 0.1–0.4 mg/kg/hours IV Sedative and anti-­tussive
Fentanyl Abstral® (Abbott Dog loading dose: 1–2 μg/kg Can cause SIADH with prolonged use
Laboratories) Dog CRI: 2–5 μg/kg/hours
Cat loading dose: 1 μg/kg/hours
Cat CRI: 1–4 μg/kg/hours
Fentanyl transdermal Duragesic® (Janssen Cat or dog < 5 kg: 25 μg patch Topical heat can increase absorption
patch Pharmaceuticals) Dog 5–10 kg: 25 μg patch Caution for abuse potential/ingestion by
Dog 10–20 kg: 50 μg patch children
Dog 20–30 kg: 75 μg patch
Dog >30 kg: 100 μg patch
Hydromorphone HCl Dog: 0.05–0.2 mg/kg IM, SQ, 0.05–0.1 mg/kg IV administration can cause vomiting
IV every q 4–6 hours
Cat: 0.05–0.1 mg/kg IM, S, 0.03–0.05 mg/kg
IV every q 3–4 hours
Methadone HCl Dog: 0.1–0.4 mg/kg IV every q 4–6 hours Tends to cause less sedation and vomiting
Dog: 0.2–2 mg/kg SQ, IM every q 4–6 hours than morphine
Cat: 0.05–0.2 mg/kg IV every q 4–6 hours
Cat: 0.1–1 mg/kg SQ, IM every q 4–6 hours
Morphine (preservative Dog: 0.25–1 mg/kg IM, SQ every q 4–6 hours IV administration must be done slowly to
free) Cat: 0.05–0.5 mg/kg IM, SQ every q 4–6 hours avoid histamine release, IV administration
Loading dose: 0.15–0.5 mg/kg IV can cause vomiting
CRI: 0.1–1 mg/kg/hour
Morphine sulfate (with Dog: 0.5–2 mg/kg IM, SQ every q 4 hours
preservative) Cat: 0.05–0.4 mg/kg IM, SQ every q 3–6 hours
Naloxone Narcan® (DuPont Pharma) Opioid reversal: 0.002–0.2 mg/kg IM, IV, SQ May need to be repeated after 20–30 minutes
as required
(Continued)

0005276422.INDD 11 04-15-2022 07:54:22


Table 1.3 (Continued)

Generic drug Brand (manufacturer) Dose Comment

Oxymorphone Numorphan® (Endo Labs) Dog: 0.02–0.2 mg/kg IV every q 1–4 hours
Dog: 0.05–0.2 mg/kg IM, SQ every q 2–6 hours
Cat: 0.01–0.05 mg/kg IV every q 2–4 hours
Lidocaine:
Lidocaine 1% Dog loading dose: 1–2 mg/kg IV Controversial for IV use in cats
preservative free Dog CRI: 20–80 μg/kg/minute
NMDA antagonists:
Ketamine KetaFlo® (Abbott Sedation: 2–10 mg/kg IV, IM Caution with hypertension, heart disease
Laboratories) Loading dose: 0.5–1 mg/kg IV Controversial in head trauma, increased
Ketaset® (Fort Dodge CRI: 0.1–0.6 mg/kg/hour ICP/IOP, renal disease (cats)
Animal Health)
Vetamine®
(Schering-­Plough)
Alpha-­2 Adrenergics
Dexmedetomidine HCl Dexdomitor® (Pfizer) Sedation: 1–10 μg/kg IV, IM Caution with cardiovascular disease or
Loading dose: 0.5–1 μg/kg IV instability
CRI: 0.25–3 μg/kg/hour
Atipamezole Antisedan® (Pfizer) Alpha-­2 Adrenergic reversal: 0.05–0.2 mg/ Same volume as dexmedetomidine given IM
kg IV, IM
Benzodiazepines:
Midazolam 0.1–0.5 mg/kg IM, IV
CRI: 0.1–0.5 mg/kg/hour
Diazepam 0.1–0.5 mg/kg IV Propylene glycol vehicle; avoid prolonged IV
CRI: 0.1–0.5 mg/kg/hour use or IM injection
Flumazenil Benzodiazepine reversal: 0.01–0.02 mg/kg May need to be repeated after 20–30 minutes
IV as required
Phenothiazines:
Acepromazine Aceproject® (Fort Dodge 0.005–0.01 mg/kg IV every 4–6 hours Caution in hypovolemia
Animal Health) 0.01–0.05 mg/kg IM, SQ every 4–6 hours Do not exceed 2 mg/kg in large dogs
Non-­steroidal anti-­inflammatory drugs:
Carprofen Rimadyl® (Pfizer) Dogs: 2–4 mg/kg IV, SQ (single dose) IV or SQ should only be given when
Dogs: 2 mg/kg PO 12 or 4 mg/kg PO once normothermic/normotensive
daily24 hours
Deracoxib Deramaxx® (Novartis) Dogs: 1–2 mg/kg/day

0005276422.INDD 12 04-15-2022 07:54:22


Generic drug Brand (manufacturer) Dose Comment

Meloxicam Metacam® (Boerhringer Dogs: 0.1–0.2 mg/kg IV, SQ (single dose) Black box warning for cats
Ingelheim) Dogs: 0.1 mg/kg PO or transmucosal once Transmucosal oral spray for dogs > 2.5 kg
OroCAM® (Abbott daily
Laboratories)
Piroxicam Feldene® (Pfizer) Dogs: 0.3 mg/kg PO once daily
Robenacoxib Onsior® (Novartis) Dogs: 2 mg/kg SQ 30 minutes before start of Do not divide/break/crush feline tablets,
surgery then every q 24 hours for a therefore, dose range in cats of 1–2.4 mg/kg
maximum of 3 days Do not use in dogs or cats less 4 months of
Dogs: 1–2 mg/kg PO once daily age
Cats: 2 mg/kg SQ 30 minutes before start of Do not use tablets in cats < 2.5 kg
surgery then every q 24 hours for a
maximum of 3 days
Cats: 1 mg/kg PO once daily for a maximum
of 3 days

Source: Adapted from Quant and Lee JA [105] and Perkowski [106].
CRI, constant rate infusion; ICP, intracranial pressure; IM, intramuscularly; IOP, intraocular pressure; IV, intravenously; NMDA, N-­methyl-­d-­aspartate; PO, per os (orally); SIADH, syndrome
of inappropriate anti-­diuretic hormone.

0005276422.INDD 13 04-15-2022 07:54:22


14 Triage and Initial Stabilization of the Emergency Small Animal Surgical Patient

particularly true in anorexic patients or those with renal hypertension and reflex bradycardia, is commonly seen in
dysfunction, which may require supplementation with patients with cerebral edema, hemorrhage, skull fractures,
potassium and/or phosphorus. Additionally, as many flu- and intracranial masses. To maintain cerebral perfusion
ids used in veterinary medicine are designed as “replace- pressure (CPP) in the face of intracranial hypertension,
ment” and not “maintenance” fluids, sodium values may arterial blood pressure (MAP) is increased, since CPP
increase in patients receiving prolonged intravenous fluid equals MAP minus ICP. Pressure receptors in the aortic
therapy, particularly in patients with continued free water arch and carotid bodies trigger a decreased heart rate in
loss, such as renal, gastrointestinal, skin, and respiratory response to the increase systemic blood pressure. Treatment
loss. Fluids with lower sodium concentrations such as of intracranial hypertension is imperative and is accom-
Normosol-­M, 0.45% NaCl, and dextrose 5% in water (D5W) plished with a combination of patient positioning and
may be necessary to prevent or manage hypernatremia pharmacologic intervention with mannitol or hypertonic
associated with prolonged fluid therapy and/or concurrent saline. If there is any evidence of or concern for head
hypotonic fluid losses. trauma or ICP, the patient’s head should be elevated
Fluid therapy in the burned veterinary patient requires 15–30 degrees using a flat board or other rigid surface.
special consideration, especially with respect to percentage Pillows should not be used to elevate the patient’s head, as
of total body surface area affected. For information on fluid this can cause compression of the jugular vein(s), which
therapy for the burned patient, see Chapter 53. impairs cerebral venous outflow. Jugular venipuncture
Regardless of fluid type and rate used to treat shock and should also be avoided. Mannitol (0.25–1 g/kg IV over
or dehydration, frequent patient reassessment is critical. 15–20 minutes) is an effective osmotic diuretic to decrease
General recommendations for patient re-­evaluation are intravascular volume and facilitate fluid movement from
listed in Table 1.2. the central nervous tissue, as with cerebral edema. The
resultant diuresis will lead to dehydration if mannitol
administration is not followed by intravenous fluid therapy
­Neurologic Assessment in patients that cannot or will not drink. Hypertonic saline
(7.5%, 3–5 ml/kg IV over 15–20 minutes in dogs, 2–3 ml/kg
Initial neurologic assessment often occurs concurrently IV over 15–20 minutes in cats) is also effective for treating
with respiratory and cardiovascular triage. Patients with intracranial hypertension secondary to cerebral edema and
normal consciousness are alert and aware of their environ- works via free water osmotic shifting out of the tissue and
ment. Obtunded patients have decreased responsiveness into the hypertonic intravascular space created by the
that can vary in severity. Stuporous patients are only increased sodium load [78]. Concentrated (21%) sodium
responsive to noxious or excessive stimuli, whereas coma- chloride can be combined with 0.9% saline to create a 7.5%
tose patients do not respond to any stimuli. Decreased cer- solution by mixing 17 ml of 21% saline with 43 ml of crys-
ebral perfusion and oxygenation from hypovolemic, talloid. The use of corticosteroids is contraindicated in
hypoxemic, distributive, and cardiogenic shock can have patients with head trauma, as they can contribute to gas-
profound effects on mentation, so the patient’s initial neu- trointestinal ulceration, especially after an episode of
rologic assessment must be made with patient’s global per- hypoperfusion, and precipitate hyperglycemia, which has
fusion status in mind. In both veterinary and human been associated with a worsened neurologic injury in vet-
patients with traumatic brain injury, most have also sus- erinary patients [79].
tained concurrent injuries to other major body systems Veterinary patients with any history of or concern for cer-
that can have secondary neurologic consequences [76, 77]. vical or spinal trauma should be secured to a backboard until
Hypoglycemia (metabolic shock) can also lead to decreased a complete assessment of injuries is performed. In trauma
mentation and must be treated before an accurate neuro- patients, assessment of cranial nerves, visual examination for
logic examination can be performed. As with other body external signs of head trauma, and any abnormalities of body
systems, frequent neurologic reassessment is imperative. position, spinal reflexes, and the presence or absence of pain
While performing the neurologic evaluation, until ade- sensation should be determined before administration of
quate oxygenation is confirmed, supplemental oxygen drugs that may impact interpretation of findings, including
should be provided by mask, flow by, or placement of the analgesics and atropine. However, assessment for the pres-
patient in an oxygen cage. In patients with head trauma, ence of a head tilt, physiologic nystagmus, and postural
nasal prongs or nasal oxygen catheter are avoided to reflexes should only be performed if it is safe to move the
decrease the risk of sneezing, which can increase their patient’s neck and limbs. Body position changes that can be
intracranial pressure (ICP). Heart rate and blood pressure seen in patients with trauma, spinal cord lesions, or intracra-
values can also provide important insight about the pres- nial disease include Schiff–Sherrington (forelimb extensor
ence of increased ICP. The Cushing’s reflex, which is rigidity and hindlimb flaccidity associated with a T2-­L4
­Urinary Assessmen  15

spinal cord lesion), decerebrate rigidity (neck extension, pigmenturia, and recent trauma can raise suspicion for
hyperextension of all four limbs, and decreased conscious- urinary tract dysfunction, however, some patients have vague
ness), and decerebellate rigidity (thoracic limb hyperexten- and non-­specific historical signs. For example, many male
sion, variable changes in the pelvic limbs, and appropriate cats with urethral obstruction present for lethargy and/or
consciousness). constipation, as many owners are unable to differentiate
A veterinary modified Glasgow Coma Scale score stranguria from tenesmus.
(mGCS) has been developed and evaluated retrospectively After respiratory, cardiovascular, and neurologic assess-
for assessing the severity of neurologic injury [80]. Scores ments have been performed and treatment of urgent
are determined after assessment of level of consciousness, abnormalities initiated, assessment of the urinary tract can
cranial nerve function, and motor function with higher be performed. Palpation for a urinary bladder should be
scores (15–18) being associated with a better prognosis performed in all patients to assess for urethral obstruction,
than lower scores (3–8 for grave prognosis and 9–14 for which causes a large, firm, painful, bladder that is unable
poor to guarded). Scoring and exact prognostication should to be expressed. Palpation of a bladder in trauma patients
be performed with caution however, since the mGCS has does not rule out injury and leakage, as small tears may not
not been prospectively evaluated and patient scores may completely decompress the bladder. Additionally, lack of a
improve with therapeutic intervention and time. In a study palpable bladder is not always synonymous with rupture,
of injured dogs and cats for whom the mGCS was used as as the bladder may not be palpable due to small size from
part of trauma scoring for a veterinary trauma database, dehydration, recent expression, or anuric or oliguric renal
mGCS scoring system corresponded with outcome in dogs failure. Ultrasound is a useful tool in the emergency room,
and cats with known head trauma [81, 82]. especially for urinary tract assessment. A standardized
For patients with deficits in conscious proprioception, technique for abdominal ultrasonographic assessment in
motor function, and pain sensation, spinal reflexes should veterinary trauma patients has been created and validated.
be used for neurolocalization of spinal cord dysfunction to In human medicine, a similar technique has largely
segments C1–C5, C6–T2, T3–L3, L4–S1, and S1–S3. replaced the need for diagnostic peritoneal lavage in blunt
Common causes of spinal cord disease in veterinary abdominal trauma patients. Focused assessment sonogra-
patients include intervertebral disc disease (IVDD), phy for trauma (FAST) has been validated to determine
trauma, neoplasia, vascular events, and infectious/inflam- whether free fluid is present in the abdominal cavity after
matory processes. Surgical intervention could be indicated trauma [83]. It is a more sensitive diagnostic for free fluid
for traumatic, neoplastic, and IVDD, especially those con- than the presence of a palpable fluid wave, which requires
ditions resulting in neurologic dysfunction. Patients with at least 40 ml/kg of fluid within the peritoneal cavity. Using
rapidly progressive neurologic changes and loss of deep ultrasound in transverse and longitudinal planes, and the
pain may require emergency diagnostic imaging and surgi- patient in lateral recumbency, the abdomen is evaluated at
cal intervention, especially if IVDD is the cause. Patients the gravity dependent and independent flanks (in the
with cervical lesions (C1–C5) are at risk of ventilatory fail- region of the kidneys), over the bladder, and below the
ure due to phrenic and intercostal nerve involvement, par- xiphoid. If needed, ultrasound guidance or blind abdomi-
ticularly after surgical decompression, as there will be the nocentesis via a one-­ or four-­quadrant closed needle/
added impact of secondary surgical swelling and hemor- syringe technique can be used to collect any free fluid. In
rhage. Respiratory pattern, effort, and objective measures many patients with significant dehydration, free fluid may
of ventilation (PvCO2 or PaCO2) should be monitored very not be present in the peritoneum initially, and serial moni-
closely. Changes in oxygenation (PaO2 and SpO2) as a result toring as the patient is rehydrated should be performed.
of ventilatory failure may be late findings and should not Once fluid is obtained, PCV/TS, glucose, lactate, creati-
be the sole determinant of effective ventilation and respira- nine, potassium, cytology, and culture can help determine
tion. Mechanical ventilation may be necessary in patients the etiology of the effusion. In dogs, a fluid to blood creati-
with cervical lesions and should be anticipated in all post- nine ratio of greater than 2 : 1, and fluid to blood potassium
operative patients with cervical neurolocalization. ratio of greater than 1.4 : 1 is supportive of a diagnosis of
uroabdomen [84]. In cats, a fluid to blood creatinine ratio
of 2 : 1 and fluid to blood potassium ratio of 1.9 : 1 is sup-
­Urinary Assessment portive of a diagnosis of uroperitoneum [85, 86].
Initial bloodwork may reveal azotemia, which could be
Many injuries and abnormalities of the urinary tract are due to prerenal, renal, or post-­renal causes. Assessment of
not readily apparent on initial triage and physical examina- urine specific gravity in conjunction with azotemia and
tion. Historical information from the pet owner regarding PCV/TS can help to determine the etiology, but it may not
changes in water consumption, urine production, stranguria, be feasible or safe to obtain a urine sample during triage
16 Triage and Initial Stabilization of the Emergency Small Animal Surgical Patient

and before initiation of fluid therapy. This is especially true


of female dogs and cats, and male cats where sedation for
catheter placement is often necessary. Cystocentesis may
be contraindicated in patients where there is any concern
for coagulopathy, thrombocytopenia, or thrombopathia.
An assessment of prerenal azotemia is therefore often
made on physical examination findings and evidence of
hemoconcentation without concurrent urine specific grav-
ity. In these cases, improvement or resolution of azotemia
in response to fluid therapy and rehydration helps to sup-
port the diagnosis of prerenal azotemia. For stabilization of
the uremic patient, please refer to Chapter 22. (a)
Urine drainage techniques can be useful for exact urine
production quantification and management of urine
leakage. Urinary catheterization can be very helpful in
patients with urinary trauma to document appropriate
urine production, maintain bladder decompression in
patients for whom concern over bladder or urethral
trauma exists, and for patient comfort and ease of man-
agement in non-­ambulatory patients. However, this use is
controversial owing to the risk of ascending urinary tract
infections, particularly in a hospital setting. Urinary cath-
eterization is easy to perform in most male dogs without
sedation, but placement in female dogs and male and (b)
female cats requires heavy sedation and can be more tech-
Figure 1.7 Distal end of a locking loop, or pigtail, catheter. The
nically challenging, especially in small female dogs catheter coil is straightened over a stylet and trocar for
(Figure 1.6). Urinary drainage with locking loop or “pig- placement (a), and then locked into the loop configuration using
tail” catheters in the peritoneum can be helpful to facili- the suture upon removal of the stylet and trocar (b).
tate urine drainage and permit patient stabilization in
preparation for surgical intervention when a uroabdomen nephrostomy tube when there is a ureteral obstruction or
is present (Figure 1.7a,b). The same catheters can be used ureteral injury [86, 87].
in the bladder for decompression when transurethral
catheterization is not possible, and in the renal pelvis as a
­Vascular Access

Vascular access is critical in emergency patients, especially


those undergoing surgery. Peripheral venous catheters are
most commonly used, since they are relatively inexpensive,
widely available, and can be placed quickly in emergency
situations. However, they do not usually allow for repeated
blood sample collection and are not appropriate for hyper-
osmolar fluids, including total or partial parenteral nutri-
tion (TPN, PPN). These catheters can also be dislodged,
soiled, and if a small-­bore catheter is used, they will not
permit rapid administration of large volumes of fluids or
medications. Vessels commonly used for peripheral cathe-
ters in small animal patients are the cephalic or accessory
cephalic veins, lateral saphenous vein, or distance branches
the medial saphenous vein [88]. Clipping of the hair and
Figure 1.6 Cadaveric dissection of the urethral papilla in a
sterile preparation of the site are preferred, but not possible
female dog, which is the major landmark used when performing in all emergency situations. Failure to adequately prepare
urinary catheterization. the skin prior to catheter placement has been associated
­Vascular Acces  17

with increased positive bacterial cultures compared with ischium and greater tubercle of the humerus [88, 92], with
those that were aseptically placed [89]. In severely hypov- the trochanteric fossa and tibia used most commonly.
olemic patients for whom these vessels cannot be cannu- Contraindications of placement of an IO catheter include
lated percutaneously, surgical cut-­down to facilitate fracture of the bone intended for cannulation, pneumatic
vascular access can be performed quickly and safely in bones in birds, and evidence of infection near the intended
small animal patients. Catheters placed without adequate catheter site. Bone growth is not impacted by IO catheteri-
skin preparation and sterile technique or with an emer- zation [95]. IO catheterization can be achieved with a vari-
gency surgical cut-­down procedure should be considered ety of techniques, including standard hypodermic needles
temporary and removed once additional vascular access is and spinal needles, IO infusion needles, a spring-­loaded
obtained and the patient is more stable to prevent infection. penetration injection gun (Vet B.I.G Bone Injection Gun
When repeated blood sample collection is anticipated, or (15-­G), WaisMed Ltd, Houston, TX) and an automatic
for administration of multiple fluids or medications simulta- rotary insertion drill (EZ-­IO (15-­G Pediatric Needle Set),
neously, including TPN or PPN, central venous catheteriza- Vidacare Corporation, San Antonio, TX.). In a cat cadav-
tion should be considered. Central catheters can have up to eric study comparing these devices, the injection gun was
four lumens, which is convenient for concurrent adminis- found to be faster and easier to use, but there were no dif-
tration of several intravenous therapies. If administration of ferences detected between insertion site (humerus or tibia),
parenteral nutrition is anticipated, one lumen should be complications or success between the injection gun, rotary
reserved specifically for this use and labeled accordingly. drill or manual IO catheter [96].
Central catheters are generally placed in larger vessels, Direct arterial blood pressure measurement should be
including the jugular vein and medial and lateral saphenous considered in any hemodynamically unstable patient. It
veins. They are most commonly placed using the Seldinger, allows for continuous, accurate pressure determination in
or over the wire, technique (Video 1.3). Surgical cut-­down the face of hypotension, hypertension, and arrhythmias.
for vascular access for Seldinger technique or venotomy and Indwelling arterial catheters can also be used to obtain
through the needle (BD Intracath, Argon Medical Devices, blood samples, particularly for arterial blood gas analysis.
Franklin Lakes, NJ) catheters can also be used. Regardless of Arteries generally accessible for percutaneous placement
the technique for placement, since a large vessel is being of an arterial catheter include the dorsal metatarsal artery
accessed, adequacy of primary and secondary hemostasis (most commonly used), the coccygeal artery in the tail, the
must be confirmed prior to placement [88, 90]. Strict aseptic auricular artery in the dorsal pinna, the femoral artery, and
technique must be followed. It is also important to have ade- the radial artery [88, 97–99]. Maintenance of arterial cath-
quate patient restraint, which generally requires sedation or eters in all locations, except for the dorsal metatarsal artery,
anesthesia, since maintenance of positioning and discom- is difficult in mobile patients and is generally reserved for
fort of vessel dilation is not well tolerated by many awake use in sedated or anesthetized patients. Femoral, dorsal
patients. The catheter is secured to the skin with sutures, metatarsal, and coccygeal artery catheters can also become
and the insertion site covered by a protective wrap. contaminated with urine and/or feces, so consideration of
these issues is important prior to catheter placement. Cats
Video 1.3 Placement of a jugular multi-­lumen catheter
tend to have poor collateral circulation. It is not recom-
using Seldinger technique after a peripheral catheter has
mended to leave arterial catheters in cats for longer than
been placed in the jugular vein.
six to eight hours because of concern for ischemic injury to
When intravenous catheterization is not possible, which the tissues distal to the catheter [97]. Contraindications for
is often the case in neonatal and small pediatric patients, arterial catheterization include lack of close monitoring
intraosseous (IO) catheterization provides a rapid, safe capabilities, thrombocytopenia, thrombocytopathia, and
method for delivery of fluid therapy and medications. This coagulopathy.
is because the capillary network within the marrow cavity Once the course of the artery is determined by palpation
is in direct communication with the nutrient and emissary and the site is aseptically prepared, an over-­the-­needle cath-
veins that drain into the central circulation. Crystalloids, eter is used to puncture the artery from an angle of
colloids, blood products and medications, including those 15–30 degrees above the vessel. Pulsatile blood flow will be
for cardiopulmonary resuscitation, can be administered via observed in the hub of the catheter upon successful arterial
the IO route and can be absorbed rapidly enough to be cannulization. If percutaneous placement of an arterial cath-
effective for the treatment of hypovolemic shock and car- eter is not possible, access to the dorsal metatarsal or femoral
diopulmonary arrest [91–96]. Sites commonly used for IO artery can be achieved with a surgical cut-­down or ultra-
catheterization include the trochanteric fossa of the femur, sound guidance [98]. Care must be taken to avoid damaging
proximal tibia, tibial tuberosity, wing of the ileum, the the artery, femoral vein, or sciatic nerve during the initial
18 Triage and Initial Stabilization of the Emergency Small Animal Surgical Patient

skin incision and approach to the femoral artery. The cathe- Opioids are often the first choice of analgesia in critical
ter is secured with tape and/or sutures depending on the veterinary patients as they have rapid onset of action, can
catheter type placed and placement method use. Once the be titrated to an individual’s needs, are reversible, and are
arterial catheter is in place, it can be used to collect arterial cardiopulmonary sparing medications. They can be used
blood samples after an adequate pre-­sample of blood is taken alone or in combination with other analgesics and anxio-
(generally 3–6 ml of blood into syringes with small amounts lytics and administered as a bolus or constant rate infusion
of heparinized saline). Clear labeling of an arterial catheter is (CRI). Common adverse effects include initial excitatory
imperative to ensure that only heparinized saline is injected phase, nausea, vomiting, bradycardia, decreased gastroin-
into the artery. Blood collected from the artery as a pre-­ testinal motility, and respiratory depression at high doses.
sample and medications should never be injected into the Cats tend to be more prone to developing an excitatory
arterial catheter. The catheter can also be connected to non-­ period, so the dose of opioids in cats is generally half the
compliant tubing with heparinized saline and a pressure canine dose. Rapid intravenous administration of mor-
transducer for continuous arterial blood pressure monitoring. phine or meperidine can cause histamine release, vasodila-
tion, and hypotension. If adverse effects result from opioid
administration, naloxone is a pure antagonist for opioid
­Analgesia, Sedation and Anxiolytics reversal. Naloxone will reverse both the positive and nega-
tive effects of opioids, which may result in pain, excite-
The need for sedation and anxiolytics in the stressed or scared ment, and agitation. Fentanyl transdermal patches are
veterinary patient and timely analgesics for those in pain effective ways to provide potent analgesia in an outpatient
cannot be overstated. This is particularly true in patients setting, but they can become displaced or ingested, and
needing emergency surgery, as many conditions requiring have the potential for misuse and abuse by owners, includ-
emergency surgical intervention create significant discom- ing ingestion by small children.
fort or pain. Anxiety and stress are present in many patients Lidocaine is effective as a local anesthetic and epidural
with respiratory compromise, especially those with upper analgesic. It can also be administered as an intravenous
airway obstruction, and should be addressed immediately to CRI, generally combined with an opioid such as morphine
provide relief for the patient and more accurate patient or fentanyl, with or without the addition of a ketamine
assessment. As with other body systems, frequent reassess- CRI. When given intravenously, it should be used cau-
ment is necessary to ensure adequate analgesia and patient tiously and titrated carefully in cardiovascularly unstable
wellbeing. Pain can be challenging to assess accurately in patients, as it can cause cardiac arrhythmias, tachycardia,
hospitalized feline patients, as they tend to be quieter and and seizures. This is especially true in cats, and some
more reserved than canine patients [100, 101]. debate exists as to whether this medication should be given
If a patient is assessed to be in pain, analgesics should intravenously to cats for analgesia.
be administered as soon as possible. It is not appropri- N-­methyl-­d-­aspartate (NMDA) antagonists such as keta-
ate to withhold analgesia because of concerns about mine are very effective, owing to their multiple sites of
creating cardiovascular or respiratory instability or action and effects, including analgesia, neuroprotection,
masking changes in patient status. When titrated doses and sedation. Ketamine has limited cardiopulmonary
of cardiovascular sparing analgesics and anxiolytics are depression but can increase cardiac output and myocardial
used, primary cardiovascular or respiratory depression oxygen consumption, and it can cause muscle tremor activ-
should not result. Instead, if hypotension or respiratory ity. Controversy exists over its use in patients with head
changes are seen after drug administration, it is more trauma as there is concern that ketamine contributes to
likely that the patient’s cardiopulmonary instability was increased ICP. It should be used with caution in patients
uncovered by relief of pain-­induced tachycardia, with hypertension and cardiovascular disease. In cats, it is
tachypnea, and catecholamine release with secondary renally excreted so consideration for renal function with
vasoconstriction [100]. use and dose should be given.
Multimodal analgesia is preferred in many patients, espe- Alpha 2 (α2) adrenergic agonists, such as dexmedeto-
cially those with marked pain, as the complexity of pain midine, bind central α2 receptors to result in sedation,
pathways often renders single agent therapy ineffective, irre- muscle relaxation, and analgesia. Additional effects
spective of dose escalation [101]. The addition of local anal- include vasoconstriction, reflex bradycardia, and diure-
gesics is often beneficial to decrease the systemic dose. The sis. Dexmedetomidine should be used cautiously in criti-
addition of anxiolytics may also be helpful to decrease cally ill patients and should be reserved only for patients
the stress and agitation associated with hospitalization, without cardiovascular disease or compromise. At low
recumbency, and activity restriction present in many surgical doses (1–5 μg/kg IV), dexmedetomidine is synergistic
patients, and therefore decrease systemic analgesic dosing. with opioids for analgesia and can be used as a bolus or
­Analgesia, Sedation and Anxiolytic  19

CRI. Its effects can be reversed with the α2 receptor takes approximately 15 minutes to achieve maximal
antagonist atipamezole. The volume of atipamezole used effect, so this delay in onset of action should be antici-
for reversal is the same volume as the administered dex- pated in patients [101]. This is important in patients in
medetomidine. Intramuscular administration of atipam- respiratory distress, for whom this delay may not be tol-
ezole is preferred to prevent rapid drug reversal, which erated, and more rapidly acting medications should be
can cause hypotension or aggression [102–104]. selected.
Benzodiazepines are effective for mild sedation and anxi- Non-­steroidal anti-­inflammatory drugs (NSAIDs) have
olysis with minimal cardiovascular compromise. They are a limited role in treating pain and inflammation in many
commonly combined with opioids for analgesia and seda- emergency patients, especially those with gastrointesti-
tion and can decrease the dose of opioids needed to achieve nal and renal disease or cardiovascular compromise.
the desired effect. Midazolam and diazepam can be given Even NSAIDs that can be administered parenterally
intravenously, but only midazolam can be given intramus- should be used with extreme caution in patients with
cularly and is preferred for CRI therapy due to the propyl- perfusion abnormalities, as they can increase the risk of
ene glycol vehicle of diazepam. Reversal of both agents can gastrointestinal ulceration, hepatic insult, and kidney
be accomplished with intravenous dosing of flumazenil. injury. NSAIDs are generally not recommended for cats,
Phenothiazines, such as acepromazine, provide no unless given as a single dose in healthy, hydrated, and
analgesia but are potent anxiolytics in veterinary medi- normovolemic cats. An NSAID designed for safer use in
cine. They must be used cautiously in cardiovascularly cats (robenacoxib) is available, but extensive clinical
unstable patients as they can cause profound vasodila- experience is lacking. However, postoperatively, when
tion and hypotension. They are especially useful in perfusion is restored and normalized, many surgical
patients with respiratory distress, particularly upper air- patients benefit from control of inflammation and the
way obstruction. However, intravenous acepromazine analgesia achieved with NSAID therapy.
20

Operating Room Nursing Tips for Emergency Surgical Procedures


Cami Elliott1, Michelle Capps2, and Michael McCallum2
1
Nashville Veterinary Specialists and Animal Emergency, Nashville, TN, USA
2
University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA, USA

­Gastrointestinal System Rectal Prolapse


A prolapsed rectum can be reduced and retained in its nor-
Most gastrointestinal procedures can be performed with mal position by applying a purse-­string suture around the
routine instruments, with a few additional items supplied circumference of the anus. Nylon suture (3-­0) on a straight
as needed. It may be beneficial to establish a routine soft needle is ideal for this purpose. Prior to reducing the pro-
tissue instrument set, such as the one listed in Box 2.1. lapse, white granulated sugar can be applied to the pro-
Unless otherwise noted, patients will be placed in dorsal lapsed tissue to alleviate some of the tissue edema. A
recumbency for gastrointestinal surgeries. It is also of ben- routine soft tissue instrument set should be sufficient, with
efit to have a variety of sizes of containers and formalin the addition of a culturette due to the location of the sur-
available for biopsies taken during gastrointestinal surgery. gery and concern for contamination. The patient is posi-
tioned in sternal recumbency in Trendelenburg position if
Esophageal Surgery the surgery table tilts. Alternatively, a rectal stand to ele-
A lodged esophageal foreign body is the most common vate the caudal end of the patient to approximately shoul-
indication for esophageal surgery and the location of the der level of the surgeon can be used.
foreign body will determine the approach. The cervical
esophagus is exposed through a ventral midline approach Hemoabdomen
and the thoracic esophagus is approached via a lateral thor-
When preparing an operating room for a patient with a
acotomy. Recommended instruments will depend on the
hemoabdomen, the first step upon entering the abdomen
approach chosen and are listed in Box 2.2.
will be to remove the accumulated blood to visualize the
source of hemorrhage. See Figure 2.1 for suction and lav-
Gastrointestinal Foreign Bodies
age instrumentation. In addition to the routine soft tissue
When preparing the operating room for a gastric or intesti- instrument set, sterile suction tubing to connect to wall-­
nal foreign body surgery the technician should keep in mounted or portable suction and additional suction can-
mind that a gastrotomy, enterotomy, or intestinal resection isters should be available, depending on the volume of
and anastomosis could be performed. To prepare for these fluid to be removed. A Balfour retractor will be helpful
events, consider that preventing the spillage of ingesta into for abdominal wall retraction while the surgeon com-
the abdominal cavity is of key concern. Sterile lap sponges, pletes a thorough examination of the abdomen to iden-
lavage, and suction are crucial in preventing contamina- tify the source of hemorrhage. Monopolar cautery and a
tion and peritonitis. Portable suction machines are availa- bipolar vessel-­sealing device (LigaSure™) are helpful in
ble if the operating room is not equipped with wall suction controlling hemorrhage and expediting the surgical
(Box 2.2, Figures 2.1 and 2.2). procedure.

Small Animal Surgical Emergencies, Second Edition. Edited by Lillian R. Aronson.


© 2022 John Wiley & Sons, Inc. Published 2022 by John Wiley & Sons, Inc.
Companion website: www.wiley.com/go/aronson/surgical
­Gastrointestinal Syste  21

Box 2.1 Routine Soft Tissue Instrument Set


●● 3 straight and 1 curved carmalt forceps
●● 4 Allis tissue forceps
●● 8 Kelley forceps
●● 6 mosquito forceps
●● 4 large Backhaus towel clamps
●● 10 small Backhaus towel clamps
●● 2 needle drivers
●● Poole suction tip
●● 2 Army Navy retractors
●● 2 Parker retractors
●● 2 basins
●● #3, #4 scalpel handles
●● Debakey thumb forceps
●● Brown–Adson thumb forceps Figure 2.1 Balfour retractor (bottom left), Poole suction tip (top
left), sterile suction tubing (bottom right), and suction canister
●● Rat-­tooth forceps
(top right) for abdominal lavage.
●● 20 4 × 4 gauze sponges

Box 2.2 Instruments for Gastrointestinal Surgery


●● Routine soft tissue instrument set
●● Balfour retractor (used for ventral midline abdominal
approach)
●● Sterile suction tubing
●● Suction canister
●● Monopolar and bipolar cautery (bipolar cautery
should be available for esophageal surgery)
●● Doyens intestinal clamps
●● Lap sponges
●● Suture for enterotomy closure
●● 25-­gauge needle and 5–10 cc syringe for performing
a leak test after enterotomy closure
●● See Box 2.12 for additional instruments necessary for
a lateral thoracotomy approach and closure.

Biliary Emergencies Figure 2.2 Portable suction machine.

In cases of biliary obstruction due to choleliths, cholecysti-


tis, pancreatitis, neoplasia or gallbladder mucocele, either Box 2.3 Instruments for Biliary Surgery
biliary duct stenting for reversible obstruction or cholecys-
Routine soft tissue instrument set
tectomy or cholecystoenterostomy for non-­reversible
●●

Monopolar and bipolar cautery


obstruction can be performed. Instruments recommended
●●

Balfour retractor
for biliary emergencies are listed in Box 2.3.
●●

●● Sterile suction tubing


●● Suction canister
Liver Lobectomy ●● Lap sponges
Culturette
For a liver lobectomy procedure in which there is potential
●●

Hemoclips® or Surgiclips™
for significant hemorrhage, a bipolar vessel sealing device
●●

± Covidien™ feeding tube and red rubber urethral


(LigaSure) can be beneficial in providing hemostasis in areas
●●

catheter (Kendall™) for common bile duct stenting


where ligations by hand would be difficult. The LigaSure
22 Operating Room Nursing Tips for Emergency Surgical Procedures

handpieces come in a variety of shapes, such as the Precise, and oxidized regenerated cellulose (Surgicel®). Both are
Impact, and Atlas. The Atlas comes in an assortment of applied to a site of hemorrhage to aid in clot formation and
lengths, diameters and with blunt and tapered tips. In addi- can be left in place indefinitely until eventually absorbed or
tion, Hemoclips, Ligaclips®, and Surgiclips are valuable removed during surgery once hemostasis is achieved. The
additions to your operating room instrumentation when instruments needed are listed in Box 2.4.
there is high risk for hemorrhage because they can be
applied quickly and securely to vessels. Removal of the
affected liver lobe can be achieved using a stapling device, ­Urinary System
such as the TA™ or GIA™ vascular stapler, which applies
two to four rows of interlaced staples. For reference, the TA Ureteral Surgery
Auto Suture vascular stapler by Covidien comes in cartridge
Instruments used for ureteral surgery are listed in Box 2.5
lengths of 30 mm, 55 mm, or 90 mm, with staple widths
and shown in Figures 2.6–2.10.
ranging from 3–4 mm and staple height ranging from
2.5–4.8 mm. For larger tissue resections, the GIA stapler
comes in cartridge lengths of 50 mm, 60 mm, 80 mm, and Urethral Surgery
90 mm, with staple widths from 3–4 mm and staple heights Perineal Urethrostomy in Cats: Patient Positioning
ranging from 2.5–4.8 mm. Other hemostatic agents include A perineal urethrostomy can be performed in sternal
Vetspon® absorbable hemostatic gelatin sponge (gel foam) recumbency with the patient’s hind legs hanging over the

Box 2.4 Instrumentation for Hepatic Surgery


●● Routine soft tissue instrument set
●● Balfour retractor
●● Monopolar cautery
●● Sterile suction tubing
●● Multiple suction canister
●● Lap sponges
●● Stapling equipment (Figure 2.3 and Table 2.1)
●● Vessel sealing device (LigaSure) (Figure 2.4)
●● Weck Hemoclips®, Ethicon Ligaclips®, or Covidien
Surgiclips® (Figure 2.5)
●● Vetspon absorbable hemostatic gelatin sponge
(Figure 2.5)
●● Oxidized regenerated cellulose (Figure 2.5)

Figure 2.3 Stapling device and cartridge options.

Table 2.1 Stapling cartridge information chart.

Staplesa
Instrument
cartridge Width × height (mm) Closed height (mm) Configuration

TA 30. 55. 90 blue 4.0 × 3.5 1.5 Double staggered row of staples
TA 30. 55. 90green 4.0 × 4.8 2.0 Double staggered row of staples
TA 30–V3 whiteb 3.0 × 2.5 1.0 3 staggered rows
GIA 50 4.0 × 4.0 1.75 4 staggered rows of staples with a cut
GIA 60 3.0 × 2.5 1.0
GIA 60 or 80 3.0 × 3.85 1.5 between 2
GIA 90 4.0 × 4.0 1.75
a
Absorbable staples: PDS and glycolic acids; stainless steel 316 l.
b
Vascular.
­Urinary Syste  23

Figure 2.5 An assortment of Hemoclip® and Ligaclip® sizes,


Figure 2.4 The bipolar vessel sealing device (LigaSure, Surgicel®, Vetspon® (Gel-­foam®), and Surgiclip® applicator
Medtronics, Dublin Ireland), comes in many size and shape (bottom). Both Gel-­foam and Surgicel are applied to a site to
options. The device aides in decreasing intraoperative and aid in clot formation and can be left in place indefinitely until
postoperative hemorrhage. eventually absorbed, or they can be removed during surgery
once hemostasis is achieved.

instruments are listed in Box 2.6 and illustrated in


Box 2.5 Instrumentation for Ureteral Surgery
Figures 2.13 and 2.14.
●● Routine soft tissue instrument set
●● Balfour retractor Retrograde Hydropulsion in Dogs
●● Sterile suction tubing Retrograde hydropulsion in dogs can be used to flush ure-
●● Suction canister thral calculi back into the urinary bladder to relieve ure-
●● Sterion® Silicone vessel loops thral obstruction prior to a cystotomy for stone retrieval.
●● Weck-­Cel® cellulose eye spears Instruments necessary for this technique are listed in
●● Microsurgical needle holders (cats and small dogs) Box 2.7.
●● Microsurgical scissors (cats and small dogs)
●● Microsurgical forceps (cats and small dogs) Urethrostomy and Urethrotomy in Dogs:
●● Bipolar cautery Patient Positioning
●● Mixter forceps The patient will be positioned in dorsal recumbency for a
●● Surgical loupes prescrotal or scrotal urethrostomy or urethrotomy and in
●● Operating microscope (recommended for cats and sternal recumbency, with the patient’s hind legs hanging
small dogs) over the end of the table and the tail retracted cranially for
●● 5-­0 to 8-­0 sutures a perineal urethrostomy or urethrotomy. Depending on the
size of the animal, magnification may be warranted.
Recommended instruments are listed in Box 2.8.

end of the table and the tail retracted cranially, as long as


Contrast Studies
there is padding to prevent obturator nerve compression. A
roll of bubble wrap can be used, or towels placed under the For some patients, the use of an iodinated contrast material
patient’s pelvis (Figure 2.11). Alternatively, the patient can (Omnipaque™) in conjunction with radiography or fluor-
be placed in dorsal recumbency with the hind limbs oscopy can provide preoperative or intraoperative informa-
retracted gently cranially (Figure 2.12). The necessary tion about the location of an obstruction, as well as the
Another random document with
no related content on Scribd:
back

You might also like