Hct hematology. T Cell Reloading After Allogeneic HCT: Enhancing Immunotherapy for AML Relapse
How does flotetuzumab impact MHC class II expression in AML cells. What role do T cell-engaging therapies play in treating AML relapse after allogeneic HCT. Why is upregulation of MHC class II molecules significant for immunotherapy effectiveness. What challenges exist in treating AML relapse following allogeneic transplantation.
The Significance of MHC Class II Upregulation in AML Immunotherapy
Acute myeloid leukemia (AML) remains a challenging disease to treat, especially when relapse occurs after allogeneic hematopoietic cell transplantation (alloHCT). Recent research has shed light on promising immunotherapeutic approaches that may enhance the body’s ability to fight AML relapse. A key finding centers on the upregulation of major histocompatibility complex (MHC) class II molecules on AML cells through interferon-gamma (IFNγ) signaling.
Why is MHC class II upregulation important? MHC class II molecules play a crucial role in presenting antigens to T cells, thereby activating the immune response against cancer cells. When AML cells express higher levels of MHC class II, they become more visible to the immune system, potentially making them more susceptible to attack by T cells.
Mechanisms of MHC Class II Upregulation
Research has shown that T cell-engaging therapies, such as the dual-affinity retargeting (DART) compound flotetuzumab (FLZ) and chimeric antigen receptor T (CAR-T) cells targeting AML-associated antigens, can induce the upregulation of MHC class II molecules. This occurs through the following mechanism:
- T cell-engaging therapies activate T cells to target AML cells
- Activated T cells release interferon-gamma (IFNγ)
- IFNγ signaling triggers upregulation of MHC class II molecules on AML cells
- Increased MHC class II expression enhances antigen presentation and immune recognition
Flotetuzumab: A Promising T Cell-Engaging Therapy for AML
Flotetuzumab (FLZ) has emerged as a promising immunotherapeutic agent for treating AML relapse after alloHCT. As a CD123-targeting DART molecule, FLZ works by bringing T cells into close proximity with AML cells, facilitating immune-mediated killing.
How does flotetuzumab enhance the immune response against AML? The mechanism involves:
- Binding to CD123 on AML cells and CD3 on T cells
- Activating T cells to release cytokines, including IFNγ
- Inducing MHC class II upregulation on AML cells through IFNγ signaling
- Increasing visibility of AML cells to the immune system
This dual action of direct T cell engagement and enhancement of antigen presentation makes flotetuzumab a potentially powerful tool in combating AML relapse.
CAR-T Cell Therapy: Another Avenue for AML Immunotherapy
Chimeric antigen receptor T (CAR-T) cell therapy represents another innovative approach to treating AML relapse. CAR-T cells are engineered to target specific antigens on AML cells, such as CD123, CD33, and CD371.
How do CAR-T cells contribute to MHC class II upregulation? Similar to flotetuzumab, CAR-T cells targeting AML-associated antigens can:
- Directly engage and kill AML cells
- Release IFNγ upon activation
- Induce paracrine IFNγ signaling in the tumor microenvironment
- Trigger upregulation of MHC class II molecules on surviving AML cells
This mechanism suggests that CAR-T cell therapy may not only directly eliminate AML cells but also enhance the visibility of remaining leukemia cells to the immune system, potentially leading to a more robust and sustained anti-leukemic response.
Challenges in Treating AML Relapse After Allogeneic HCT
Despite the potent graft-versus-leukemia (GVL) effects mediated by donor-derived cells in allogeneic HCT, AML relapse remains a significant challenge. Several factors contribute to the difficulty in treating post-transplant relapse:
Immune Evasion Mechanisms
AML cells can develop various strategies to evade immune recognition and attack. These may include:
- Downregulation of MHC molecules
- Expression of immune checkpoint molecules
- Induction of peripheral tolerance
- Loss of mismatched human leukocyte antigen haplotypes
Genetic Instability
The inherent genetic instability of AML progenitor cells can lead to the emergence of resistant clones that are less susceptible to immune-mediated killing. This genetic plasticity allows AML cells to adapt to the selective pressures imposed by allo-reactive effector cells.
Limited Treatment Options
Conventional treatments for AML relapse after alloHCT often have limited efficacy and may be associated with significant toxicities. This underscores the need for novel therapeutic approaches that can effectively target resistant AML cells while minimizing harm to healthy tissues.
The Promise of Combining Immunotherapies for AML Relapse
The findings on MHC class II upregulation by T cell-engaging therapies open up exciting possibilities for combination treatment strategies. By leveraging the complementary mechanisms of different immunotherapeutic approaches, it may be possible to create more effective and durable responses in patients with relapsed AML.
Potential Combination Strategies
- Flotetuzumab + CAR-T cells: Combining these therapies could lead to enhanced T cell activation and more robust MHC class II upregulation.
- Immunotherapy + Epigenetic modifiers: Drugs that influence gene expression could potentially further enhance MHC class II expression on AML cells.
- T cell-engaging therapies + Checkpoint inhibitors: This combination might help overcome immune suppression mechanisms and boost the overall anti-leukemic response.
By strategically combining these approaches, clinicians may be able to create a more hostile environment for AML cells, making it harder for them to evade immune recognition and elimination.
Translating Research Findings into Clinical Practice
The promising results observed with flotetuzumab and CAR-T cell therapies in preclinical studies and early clinical trials raise important questions about how to best translate these findings into effective treatments for patients with relapsed AML after alloHCT.
Key Considerations for Clinical Implementation
- Patient selection: Identifying which patients are most likely to benefit from T cell-engaging therapies based on their disease characteristics and immune profile.
- Timing of intervention: Determining the optimal time to initiate immunotherapy after alloHCT to maximize efficacy while minimizing the risk of graft-versus-host disease.
- Monitoring and biomarkers: Developing robust methods to track MHC class II expression and other relevant biomarkers to guide treatment decisions and assess response.
- Managing toxicities: Implementing strategies to mitigate potential side effects associated with T cell-engaging therapies, such as cytokine release syndrome.
As research in this area continues to evolve, it will be crucial to design well-controlled clinical trials that can definitively establish the efficacy and safety of these novel immunotherapeutic approaches in the post-alloHCT setting.
Future Directions in AML Immunotherapy Research
The discovery of MHC class II upregulation as a mechanism of action for T cell-engaging therapies opens up numerous avenues for future research in AML immunotherapy. Some promising areas of investigation include:
Enhancing MHC Class II Expression
Researchers may explore additional methods to boost MHC class II expression on AML cells, such as:
- Developing targeted therapies that directly induce MHC class II upregulation
- Investigating the role of epigenetic modifiers in enhancing antigen presentation
- Exploring combination strategies that synergistically increase MHC class II expression
Optimizing T Cell-Engaging Therapies
Future studies may focus on refining existing T cell-engaging therapies and developing new ones, with goals such as:
- Improving the specificity and affinity of bispecific antibodies like flotetuzumab
- Enhancing the persistence and efficacy of CAR-T cells in the AML setting
- Developing novel constructs that can simultaneously target multiple AML-associated antigens
Understanding Resistance Mechanisms
As these therapies move forward in clinical development, it will be crucial to investigate potential resistance mechanisms that may emerge, including:
- Alterations in IFNγ signaling pathways
- Development of escape variants with reduced target antigen expression
- Upregulation of inhibitory immune checkpoints in response to therapy
By proactively studying these resistance mechanisms, researchers can develop strategies to overcome them and maintain long-term efficacy of immunotherapeutic approaches.
Implications for Broader Cancer Immunotherapy
While the current research focuses on AML, the principles underlying MHC class II upregulation through T cell-engaging therapies may have broader implications for cancer immunotherapy as a whole. This mechanism could potentially be exploited in other hematologic malignancies and solid tumors where enhancing antigen presentation could improve immune recognition and elimination of cancer cells.
Potential Applications in Other Cancers
- Lymphomas: Investigating whether similar approaches could enhance the efficacy of CAR-T cell therapies in B-cell malignancies
- Solid tumors: Exploring the potential of T cell-engaging therapies to increase MHC class II expression and improve immunogenicity in cancers such as melanoma or lung cancer
- Combination with other immunotherapies: Studying how MHC class II upregulation might synergize with checkpoint inhibitors or cancer vaccines across various tumor types
As our understanding of the complex interplay between cancer cells and the immune system continues to grow, the insights gained from AML research may pave the way for more effective immunotherapeutic strategies across a wide range of malignancies.
In conclusion, the discovery of MHC class II upregulation as a mechanism of action for T cell-engaging therapies represents a significant advance in our understanding of AML immunotherapy. This finding not only provides a strong rationale for the use of therapies like flotetuzumab and CAR-T cells in the post-alloHCT setting but also opens up new avenues for combination strategies and future research. As we continue to unravel the complexities of the immune response to AML, we move closer to developing more effective and personalized treatment approaches for patients facing this challenging disease.
T cells reloaded after allogeneic HCT | Blood
Skip Nav Destination
IMMUNOBIOLOGY AND IMMUNOTHERAPY|
April 6, 2023
Martin Bornhäuser
Blood (2023) 141 (14): 1652–1653.
https://doi.org/10.1182/blood.2022019106
Connected Content
A related article has been published:
Flotetuzumab and other T-cell immunotherapies upregulate MHC class II expression on acute myeloid leukemia cells
-
Split-Screen -
Share
-
Tools
-
Request Permissions
-
Cite Icon
Cite
-
-
Search Site
-
Open the
PDF for in another window
Citation
Martin Bornhäuser; T cells reloaded after allogeneic HCT. Blood 2023; 141 (14): 1652–1653. doi: https://doi.org/10.1182/blood.2022019106
Download citation file:
- Ris (Zotero)
- Reference Manager
- EasyBib
- Bookends
- Mendeley
- Papers
- EndNote
- RefWorks
- BibTex
toolbar search
Subjects:
Free Research Articles
In this issue of Blood, Rimando et al suggest that T-cell–engaging immunotherapeutic approaches may be especially suited to confront or respond to or salvage relapse of acute myeloid leukemia (AML) after allogeneic hematopoietic cell transplantation (alloHCT).1 Both the dual-affinity retargeting (DART) compound flotetuzumab (FLZ) and chimeric antigen-receptor T (CART) cells against AML-associated surface antigens such as CD123, CD33, and CD371 induced upregulation of major histocompatibility complex (MHC)-class II molecules by interferon-gamma (IFNγ) signaling. This mechanism was confirmed in cell lines and primary AML cells in vitro, in xenogeneic transplant models, and in samples of patients treated with FLZ for relapsed disease. By elucidating the mode of action, the authors provide further rational for use of T cell–engaging therapies and/or CART cells in patients with relapse after alloHCT.
AlloHCT is the most effective strategy for treating suitable patients with AML, owing to the potent graft-versus-leukemia (GVL) effects mediated by donor-derived, allo-reactive effector cells. In vitro cytotoxicity studies of matched donor cells with AML blasts and adoptive transfer studies in NSG mice support the hypothesis that MHC class II molecules are major targets of GVL.2 Despite the strength of allogeneic GVL effects, a significant proportion of patients with AML relapse after alloHCT. Treatment of relapse after alloHCT is an immense challenge, and a cure is elusive in most cases.3 The exact reason for immune escape is difficult to dissect at an individual case level, but several patterns of immune evasion have been described. Peripheral tolerance-induction and expression of immune checkpoints on leukemic cells after alloHCT may contribute to the loss of antileukemic immunity resulting in frank relapse.4 Due to inherent genetic instability, AML progenitor cells, like many other cancer cells, can downregulate MHC molecules. The mode and pattern of reduced human leukocyte antigen expression are shaped mainly by the pressures induced by allo-reactive effector cells. Complete loss of mismatched human leukocyte antigen haplotypes on the genetic level has been described after haploidentical alloHCT.5 Conversely, posttranscriptional downregulation of MHC class II expression has been detected on AML blasts at relapse after allogeneic HCT. In that same study, the incubation of leukemic cells with IFNγ led to upregulation of MHC class II expression.6
This report opens up a new and exciting strategy for treatment of relapsed AML after alloHCT. The authors of this study clearly demonstrate that both the CD123-targeting DART FLZ and AML-targeting CART cells induce paracrine IFNγ and subsequent upregulation of MHC class II (see figure). This effect was observed not only in vitro but also in a xenotransplant model and in paired patient samples analyzed before and after treatment with FLZ.
View largeDownload PPT
Acute myeloid leukemia (AML) targeted by either the dual-affinity retargeting compound (DART) flotetuzumab (FLZ) or chimeric-antigen-receptor (CAR) T cells upregulate major histocompatibility complex (MHC) class II on their surface, rendering them recognizable by allogeneic effector cells in patients after allogeneic hematopoietic cell transplantation. GvL, graft-versus-leukemia; IFNγ, interferon gamma; TCR, T-cell receptor. Illustration by Helena Jambor.
View largeDownload PPT
Acute myeloid leukemia (AML) targeted by either the dual-affinity retargeting compound (DART) flotetuzumab (FLZ) or chimeric-antigen-receptor (CAR) T cells upregulate major histocompatibility complex (MHC) class II on their surface, rendering them recognizable by allogeneic effector cells in patients after allogeneic hematopoietic cell transplantation. GvL, graft-versus-leukemia; IFNγ, interferon gamma; TCR, T-cell receptor. Illustration by Helena Jambor.
Close modal
This study provides a clear rationale to test T-cell–engaging therapies in patients with relapse after alloHCT. An interesting question to answer is whether the effects of these therapies are more durable in such patients, compared to their effects in patients who have not undergone a transplantation procedure.7 This benefit is suggested by prior reports that unspecific reactivation of donor T cells—for example, after adoptive transfer—has been shown to be of limited efficacy, but durable, in a small subset of patients.8 The limitation of adoptive transfer always has been the potential induction of graft-versus-host disease (GVHD). Such an on-target, off-tumor toxicity has to be kept in mind when T cells are activated or transferred after alloHCT and MHC expression is induced, but the risk of GVHD induction, it is hoped, will be lower when the awakening of alloreactivity is targeted to AML cells. With the importance of IFNγ signaling for the observed effects, one has to be cautious when patients have received or are still on JAK-2 inhibitors (eg, ruxolitinib). However, the additional systemic administration of IFNγ might even boost the effects of T-cell–targeting therapies. Fortunately, we will not have to wait too long to get first insights into the feasibility and efficacy of such treatment strategies, as the use of both IFNγ and FZL are currently being tested in patients with relapse after alloHCT (https://clinicaltrials.gov: NCT04628338 and NCT04582864). As the response to FLZ has been shown to be related to expression of IFNγ-related genes in the nontransplant setting, a point of interest is whether similar predictive biomarkers can be identified in the post-HCT setting.9 The current paper by Rimando et al provides a convincing rationale to pursue the clinical testing of T-cell–engaging therapies and CART approaches in patients with relapse of myeloid malignancies after alloHCT.
Conflict-of-interest disclosure: M.B. reports being a scientific advisory board member of and receiving speaker honoraria from Jazz Pharmaceuticals and MSD.
1.
Rimando
JC
,
Chendamarai
E
,
Rettig
MP
, et al.
Flotetuzumab and other T-cell immunotherapies upregulate MHC class II expression on acute myeloid leukemia cells
.
Blood
.
2023
;
141
(
14
):
1718
–
1723
.
2.
Herr
W
,
Eichinger
Y
,
Beshay
J
, et al.
HLA-DPB1 mismatch alleles represent powerful leukemia rejection antigens in CD4 T-cell immunotherapy after allogeneic stem-cell transplantation
.
Leukemia
.
2017
;
31
(
2
):
434
–
445
.
3.
Zeiser
R
,
Beelen
DW
,
Bethge
W
, et al.
Biology-driven approaches to prevent and treat relapse of myeloid neoplasia after allogeneic hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2019
;
25
(
4
):
e128
–
e140
.
4.
Norde
WJ
,
Maas
F
,
Hobo
W
, et al.
PD-1/PD-L1 interactions contribute to functional T cell impairment in patients who relapse with cancer after allogeneic stem cell transplantation
.
Cancer Res
.
2011
;
71
(
15
):
5111
–
5122
.
5.
Vago
L
,
Perna
SK
,
Zanussi
M
, et al.
Loss of mismatched HLA in leukemia after stem- cell transplantation
.
N Engl J Med
.
2009
;
361
(
5
):
478
–
488
.
6.
Christopher
MJ
,
Petti
AA
,
Rettig
MP
, et al.
Immune escape of relapsed AML cells after allogeneic transplantation
.
N Engl J Med
.
2018
;
379
(
24
):
2330
–
2341
.
7.
Uy
GL
,
Aldoss
I
,
Foster
MC
, et al.
Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia
.
Blood
.
2021
;
137
(
6
):
751
–
762
.
8.
Schmid
C
,
Labopin
M
,
Nagler
A
, et al.
Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party
.
J Clin Oncol
.
2007
;
25
(
31
):
4938
–
4945
.
9.
Vadakekolathu
J
,
Minden
MD
,
Hood
T
, et al.
Immune landscapes predict chemotherapy resistance and immunotherapy response in acute myeloid leukemia
.
Sci Transl Med
.
2020
;
12
(
546
):
eaaz0463
.
Sign in via your Institution
Shared Care – Medical Learning Institute, Inc.
On-Demand
Part of the Series:
NMDP
Available: February 22, 2023 —
February 21, 2024
Credit: 0.75hr(s)
Credit Types:
CME, NCPD
Therapeutic Areas:
Hematologic Oncology, Hematology, Hematopoietic Cell Transplantation (HCT), Oncology
Specialties:
Bone Marrow Transplantation, Hematology, Oncology
Location: Internet Activity Enduring
Enroll Today
Course Description
Community hematology/oncology practices play a critical role to expand patient access to hematopoietic cell transplantation (HCT) and improve patient outcomes.
This self-guided online training course series consists of five courses designed to present the current state of the science for Hematopoietic Cell Transplantation (HCT) by following the evolution of HCT over time. The series is broken down into major misperceptions or myths about HCT and cellular therapy that are not supported by current research and clinical practice.
This is Part 5 in a 5-part series, exploring how shared and coordinated care between transplant centers and hematology/oncology practices can help overcome barriers to HCT and improve patient outcomes and experience.
Other Courses in this Series
Series:
NMDP
Expires: February 21, 2024
Credit: 1.00hr(s)
CME, NCPD
Therapeutic Areas:
Hematologic Oncology, Hematology, Hematopoietic Cell Transplantation (HCT), Oncology
Specialties:
Bone Marrow Transplantation, Hematology, Oncology
Series:
NMDP
Expires: February 21, 2024
Credit: 1. 50hr(s)
CME, NCPD
Therapeutic Areas:
Hematologic Oncology, Hematology, Hematopoietic Cell Transplantation (HCT), Oncology
Specialties:
Bone Marrow Transplantation, Hematology, Oncology
Series:
NMDP
Expires: February 21, 2024
Credit: 0.75hr(s)
CME, NCPD
Therapeutic Areas:
Hematologic Oncology, Hematology, Hematopoietic Cell Transplantation (HCT), Oncology
Specialties:
Bone Marrow Transplantation, Hematology, Oncology
Series:
NMDP
Expires: February 21, 2024
Credit: 0. 75hr(s)
CME, NCPD
Therapeutic Areas:
Hematologic Oncology, Hematology, Hematopoietic Cell Transplantation (HCT), Oncology
Specialties:
Bone Marrow Transplantation, Hematology, Oncology
Series:
NMDP
Expires: February 21, 2024
Credit: 0.50hr(s)
CME, NCPD
Therapeutic Areas:
Hematologic Oncology, Hematology, Hematopoietic Cell Transplantation (HCT), Oncology
Specialties:
Bone Marrow Transplantation, Hematology, Oncology
Other Courses That May be of Interest
Series:
RRMM
Expires: February 8, 2024
Credit: 0. 50hr(s)
AAPA, CME, CPE, NCPD
Therapeutic Areas:
Hematologic Oncology, Oncology
Specialties:
Multiple Myeloma
Expires: February 28, 2024
Credit: 1.0hr(s)
AAPA, CME, CPE, NCPD
Therapeutic Areas:
Hematologic Oncology, Hematology
Specialties:
CAR T-Cell Therapy, Lymphoma
Series:
RRMM
Expires: February 8, 2024
Credit: 0. 50hr(s)
AAPA, CME, CPE, NCPD
Therapeutic Areas:
Hematologic Oncology, Oncology
Specialties:
Multiple Myeloma
Series:
CLL/SLL
Expires: December 14, 2023
Credit: 0.30hr(s)
AAPA, CME, CPE, NCPD
Therapeutic Areas:
Hematologic Oncology, Oncology
Specialties:
Chronic Lymphocytic Leukemia (CLL), Small lymphocytic lymphoma (SLL)
Series:
NSCLC
Expires: November 28, 2023
Credit: 0. 30hr(s)
NCPD
Therapeutic Areas:
Hematologic Oncology, Oncology
Specialties:
Biomarkers, NSCLC
Series:
CLL/SLL
Expires: December 14, 2023
Credit: 0.30hr(s)
AAPA, CME, CPE, NCPD
Therapeutic Areas:
Hematologic Oncology, Oncology
Specialties:
Chronic Lymphocytic Leukemia (CLL), Small lymphocytic lymphoma (SLL)
Series:
NSCLC
Expires: November 28, 2023
Credit: 0. 15hr(s)
NCPD
Therapeutic Areas:
Hematologic Oncology, Oncology
Specialties:
Biomarkers, NSCLC
Series:
CLL/SLL
Expires: December 14, 2023
Credit: 0.30hr(s)
AAPA, CME, CPE, NCPD
Therapeutic Areas:
Hematologic Oncology, Oncology
Specialties:
Chronic Lymphocytic Leukemia (CLL), Small lymphocytic lymphoma (SLL)
View All Events
Subscribe to Mailing List
SURGERY | “RMJ” No.
7 of 04/03/1997
COST EVALUATION OF THREE ANTIMICROBIAL REGIMENS USED TO PREVENT INFECTIOUS COMPLICATIONS AFTER ABDOMINAL SURGERY
PERCUTANEOUS MITRAL COMISUROTOMY
PERCUTANEOUS TRANSLUMINAL ANGIOPLASTY FOR KAWASAKI DISEASE
COST EVALUATION OF THREE ANTIMICROBIAL REGIMENS USED TO PREVENT INFECTIOUS COMPLICATIONS AFTER ABDOMINAL SURGERY
J. Bender
AND Infectious complications after surgery pose a threat to patients, cause morbidity, affect the workload of hospital staff, and their development leads to an increase in the cost of hospital and health insurance companies for treatment.
Antibiotic prophylaxis has been beneficial over the past 20 years, and there has recently been some agreement that effective prophylaxis can be achieved with a single dose of the appropriate drug (or combination of antibiotics) at the start of surgery. The administration of a single dose reduces the likelihood of developing resistant strains of microorganisms and is cheaper than repeated antibiotics.
G. Anderson et al. from two University Hospitals in Melbourne (Australia) conducted a prospective study to evaluate the efficacy and cost of three antimicrobial regimens used to prevent infectious complications after abdominal surgery. The 1,070 patients were randomly selected to receive one of three treatment regimens consisting of ceftriaxone (1 g intravenously) or cefotaxime (1 g intravenously), both third-generation cephalosporins, or ticarcillin, a moderately broad-spectrum penicillin, in combination with clavulanic acid (3.1 g of this combination intravenously). Interventions included appendectomy, elective and emergency colorectal surgery, esophagogastroduodenal surgery, small bowel resection, cholecystectomy, and examination of the common bile duct. Patients undergoing appendectomy or colorectal surgery were given an additional 500 mg intravenous metronidazole, which is effective against anaerobic microorganisms and protozoa (Trichomonas), in cases where, after randomization, they were to receive one of the cephalosporins. Metronidazole was not added if, after randomization, the patient received ticarcillin plus clavulanic acid.
Wound infections were defined as dangerous when the hospital stay was prolonged or a second operation was required; Wound infection was considered minor if it did not affect the length of hospital stay.
The following results were obtained: the frequency of wound infections was 4.3%. Twenty-one (2%) patients developed a serious wound infection as expected, mainly after colorectal surgery, while 25 (2.3%) patients had a minor wound infection.
There was no difference in the severity of wound infections between the three groups receiving different treatment regimens; the fewest minor complications (virtually none) were observed in the ceftriaxone group, the infection being caused mainly by Staphylococcus aureus and colon dwellers such as Enterobacteriaceae and E. coli. In the frequency of postoperative infections of other localization (lungs, urinary tract) between the three groups, no differences were found.
The side effects of the antibiotics used were minimal: in the ceftriaxone group, there was 1 case of mild thrombocytopenia that lasted only 1 day; another patient treated with cefotaxime had urticaria for a short time. Considering there was no difference in infection rates between the three groups, the researchers analyzed treatment costs. They accurately took into account not only the cost of acquiring drugs, but also the cost of the necessary treatment for infectious complications that developed in any of the three groups. The cost of acquiring ceftriaxone was twice that of the other two regimens, which cost the same.
The costs of treating infectious complications were highest in the ticarcillin plus clavulanic acid group, while they were similar in the other two groups. The authors concluded that both cephalosporins are equally effective against infectious complications after abdominal surgery. Since the acquisition costs of cefotaxime are lower, it may be the drug of choice for antimicrobial prophylaxis.
References:
Anderson G, Boldiston C, Woods S, O`Brien P. A Cost-effectiveness evaluation of 3 antimicrobial regimens for the prevention of infective complications after abdominal surgery. Arch Surg 1996;131:744-8.
PERCUTANEOUS MITRAL COMISUROTOMY
N. Mazur
N. Mazur
90 005 D This method of treatment has been used for about 15 years, which made it possible to obtain immediate and medium-term results of the intervention, as well as determine the indications for it . Percutaneous commissurotomy (PCT), performed by experienced specialists, is characterized by a low risk of complications. In general, the need for this surgical intervention does not exceed 1%, the proportion of patients in whom this intervention is not possible ranges from 1 to 17%. In the case of successful commissurotomy, the area of the atrioventricular opening increases by more than 2 times (on average from 0. 9- 1 to 1.9 – 2.4 cm 2 ). The risk of hemopericardium is 0.5 – 7%, embolism – 0.5 – 5%, severe mitral regurgitation – 2 – 19%, hemodynamically insignificant atrial septal defect – 10 – 20%. Other complications (vascular, arrhythmias) are very rare.
An immediate improvement in the condition is noted with an increase in the opening area to 1.5 cm 2 or at least 25% and is manifested by a decrease in pressure in the left atrium, pulmonary artery and a decrease in pulmonary vascular resistance. Long-term 5-year follow-up showed that the harbingers of an unfavorable outcome are age, an initially high functional class of circulatory insufficiency, the value of vascular resistance of the pulmonary bed, the area of the atrioventricular opening after commissurotomy. Indications for PCT are determined by the clinical condition, anatomical changes in the valves and the experience of specialists. Intervention is usually not indicated for patients who do not present complaints, except for those who need major surgery on other organs, who want to have a baby, who are at high risk of thromboembolism or recurrent supraventricular arrhythmia. The absolute indication for PCT is the impossibility of surgical intervention. It should be preferred in the following cases: in restenosis after surgical commissurotomy; in patients with an increased risk of poor outcome during surgery; in patients with high-grade pulmonary hypertension; in patients with severely impaired left ventricular function; in patients with aortic disease in order to improve their condition before subsequent surgery on both valves; in elderly patients.
Anatomical factors that are contraindications for PCT are the following: the presence of a thrombus in the atrium, severe mitral regurgitation, severe aortic heart disease, the area of the atrioventricular orifice is more than 1.5 cm 2 . The results are significantly worse if valvular calcification is present. With massive calcification of both valves, surgical treatment is preferable. The significant influence of the specialist’s experience on the results of PCT is demonstrated by the data on the frequency of complications depending on the number of procedures performed. For example, if the experience is limited to about 100 interventions, the need for urgent surgery occurs 4 times more often than if the PCT is performed by a specialist who has already performed 1000 procedures.
Thus, PCT should be considered as an additional treatment for patients with mitral stenosis.
Literature:
Vahanian A. Percutaneous mitral comissurotomy. Eur Heart J 1996;17:1465-9.
PERcutaneous transluminal angioplasty for KAWASAKI DISEASE
S. Berreklouw
A standard therapy for severe coronary artery stenosis associated with damage due to Kawasaki disease. Long-term patency of the bypass shunt in elderly patients is satisfactory, which cannot be said for young children suffering from this disease. According to previous studies, short-term and long-term patency of shunts from the saphenous vein of the leg after 1 year is 67% and decreases to 55% after 5-9 years. years. Moreover, these shunts have a number of disadvantages: the lumen diameter of native saphenous vein grafts is too small to achieve satisfactory long-term patency in young children, and these shunts have a limited ability to expand as the child grows. Currently, bypass grafts from the anterior thoracic artery have found application in coronary artery bypass grafting due to their growth potential. However, the long-term patency of these shunts has not been determined. On the other hand, percutaneous transluminal coronary angioplasty (PTCA) as an alternative treatment for severe coronary stenosis associated with Kawasaki disease lesions is rarely performed. From 1982, at the institute where T. Ino et al. work, all patients in whom a coronary aneurysm or significant expansion is detected during two-dimensional echocardiography in the acute stage undergo coronary angiography within 6 months from the onset of Kawasaki disease. As a result, angiographic abnormalities in the coronary arteries were detected in 220 (79%) of 278 patients. The remaining 58 (21%) patients had no visible abnormalities indicative of coronary aneurysm recurrence or expansion. Of 220 patients with angiographic abnormalities, 46 had an aneurysm with or without significant stenosis, and 174 had an enlargement. Finally, 5 patients (1 female) aged 2 to 18 years (median 8 years) underwent PTCA for severe coronary artery stenosis. In the acute phase, these patients were diagnosed with Kawasaki disease, they were prescribed aspirin and prednisolone (2 patients), aspirin alone (1), aspirin in combination with high doses of gamma globulin (1). There is no information about the treatment prescribed for the fifth patient. All patients received an anticoagulant (aspirin 5–10 mg daily with or without dipyridamole 3–5 mg daily) from the time the lesion was first detected until PTCA was performed. The time from onset of Kawasaki disease to PTCA ranged from 2 to 16 years (median 6 years).
Fluoroscopy revealed calcification of the coronary artery in 2 patients. One patient had three annular calcifications in three coronary arteries. In another patient, annular calcification was consistent with an aneurysm of the left anterior descending artery. PTCA was considered successful if the degree of stenosis was less than 50% of that before dilatation. Control angiography was performed 6-12 months after the initial one.
PTCA was found to be effective in 4 out of 5 patients. Damage, which was the “target” of PTCA, in 3 patients was localized in the middle right coronary artery, and in 2 – in the proximal left anterior descending artery. In all patients, stenosis was localized proximal to the aneurysm. PTCA was successfully performed in two right-sided and two left-sided coronary stenoses. In 4 patients in whom angioplasty was effective, the degree of stenosis decreased from 84% to 33% (p < 0.005). In the fifth patient, who had annular calcification, the percentage of stenosis at PTCA remained unchanged even when the filling pressure was increased to 10 atm. Two-dimensional echocardiography did not reveal significant differences in the movement of the left ventricular wall before and after PTCA.
4 patients who underwent successful PTCA underwent follow-up angiography 6–12 months later; the study did not reveal significant restenosis and progression of stenosis. There were no significant complications that could be attributed to the performed PTCA.
PTCA was performed in these patients because the stenosis could progress and acute myocardial ischemia could develop after a few years. Criteria for performing PTCA in children with Kawasaki disease, different from those in adults, have yet to be determined. If a pronounced initial thickening is detected, then it is more advisable to use not a conventional angioplasty balloon, but a rotary destructor (rotablator) or an arterectomy catheter.
Thus, PTCA can be used as an alternative to coronary artery bypass grafting in patients with coronary artery stenosis due to Kawasaki disease. Conventional PTCA should be performed in patients younger than 8 years due to the specific histopathological features of this disease.
Literature:
Ino T, Akimoto K, Ohkubo M, Nishimoto K, Yabuta K, Takaya J, Yamaguchi H. Application of percutaneous transluminal coronary angioplasty to coronary arterial stenosis in Kawasaki disease. Circuit 1996;93:1709-15.
Efficacy of Pramistar in patients with arterial hypertension complicated by chronic cerebrovascular accident
Arterial hypertension (AH) is the main risk factor and pathogenetic mechanism for the development of chronic cerebrovascular accident (CIC), since high blood pressure (BP) is associated with changes in the structural and functional state of the walls of small penetrating arteries, leading to the development of lacunar cerebral infarctions, as well as the development occlusive processes of the main arteries. Formed and decaying atherosclerotic plaques in the main arteries with subsequent atherothrombosis, cardiac arrhythmias with intracavitary thrombosis cause the development of embolic strokes. Systemic arterial hypotension caused by heart failure or inadequate antihypertensive therapy leads to the development of ischemia in the area of vascularization of the distal segments of cerebral vessels (“beggarly perfusion” syndrome) [5-7]. Of no small importance in the development and progression of CNMC in patients with AH are microcirculation disorders caused by an increase in the functional activity of platelets, blood viscosity, endothelial dysfunction and remodeling of cerebral arteries [5-7].
Treatment of CNMC in patients with AH is currently a very urgent and difficult task. Along with adequate antihypertensive, lipid-lowering and antiplatelet therapy, correction of endothelial dysfunction, the most important component of complex treatment is nootropic therapy [1-5, 8]. According to WHO experts, nootropic drugs are drugs that have a direct activating effect on learning, improve memory and mental activity, and also increase the brain’s resistance to various aggressive influences.
The aim of the study was to evaluate the efficacy and safety of Pramistar (Pramiracetam, Berlin-Chemie, Germany) in patients with AH complicated by CNMC.
Materials and methods of research
The study involved 40 patients (25 men and 15 women) aged 45 to 75 years (mean age 60.5 ± 3.5 years) with stage II AH complicated by CICD (dyscirculatory, hypertensive encephalopathy stage II). The inclusion criteria for the study was the predominance of cognitive and autonomic disorders in the clinical picture. At the same time, these patients also had other symptoms – affective, cerebrosthenic, psychopathic, which at this stage was not the leading one.
The clinical state of the majority of patients before the start of therapy was determined by a combination of cognitive disorders of varying severity (difficulty in concentrating attention, decreased operational and long-term memory, decreased intellectual productivity and general activity, impaired speech of dysmnesic genesis) and autonomic disorders (headaches and dizziness, noise and ringing in ears, hyperesthesia to light and sounds, fatigue, irritability, internal tension and restlessness).
Exclusion criteria from the study were systolic blood pressure > 220 mmHg. and / or diastolic blood pressure > 120 mm Hg, previous stroke of any etiology, heart failure stage IIB-III, III-IV functional class, serum creatinine level > 0.15 mmol / l, decompensated diabetes mellitus, oncological, hematological , endocrinological diseases, systemic connective tissue diseases, liver and kidney diseases in the stage of decompensation, tuberculosis, chronic obstructive pulmonary diseases, mental illness.
Pramistar was studied in an open clinical trial (without placebo) on a background free from other psychotropic drugs. All patients continued to receive individual antihypertensive, lipid-lowering and antiplatelet therapy. Pramistar was prescribed at a dose of 1200 mg per day (from the first day of treatment in two doses of 600 mg). The duration of treatment was 5 weeks; clinical examinations of patients using rating scales were carried out on days 1, 21 and 35 of therapy.
The efficacy and safety of Pramistar was assessed according to the following unified rating scales: a clinical unified questionnaire based on the methodological recommendations of the Scientific and Methodological Center for Borderline Psychiatry of the State Scientific and Methodological Center for Public Health and the St. V.P. Serbsky (Russia), Hamilton anxiety severity rating scale, memorization test of 10 words with five memorization under conditions of auditory presentation (ZDS), test of memorization of semantic organized material under conditions of visual presentation, visual texts (ST), number search test with switching, black-and-white red table (CCT), adverse reactions registration cards.
Results and discussion
The severity of cognitive and autonomic disorders according to the unified clinical questionnaire decreased by 50% and 55% by the 21st day of therapy, and by 54% and 60% by the 35th day of therapy, respectively. All patients noted an increase in mental activity. Patients who continued to work reported that they became more active during the working day, coped more easily with the amount of work material, and turned to work notes and diaries more rarely. The majority of patients – 35 (87.5%) – noted a decrease in “stiffness” of thinking, an increase in understanding, analysis and assimilation of test materials and inverted speech.
Thirty-six (90%) patients had mild anxiety symptoms before treatment (mean Hamilton anxiety scale score of 18). On the 21st and 35th days of therapy, the anxiety level indicators were 13 and 10 points, respectively, which indicates the anxiolytic effect of Pramistar. In no case was there an increase in anxiety. In 32 (80%) patients, normalization of sleep and improvement in somnological parameters were noted: a decrease in the period of falling asleep, a decrease in nocturnal awakenings, and an increase in the duration of sleep.
In the study of the ability of patients to memorize and reproduce discrete verbal material using the CDS technique, a significant increase in the average number of correctly reproduced elements from 5.45 ± 1.27 to 7.36 ± 1.67 was noted – by 24.8% (p < 0, 02) by the 21st day of treatment and up to 7.85 ± 1.75 by 29.8% (p < 0.01) by the 35th day of treatment.
The study using the ST method revealed a significant improvement in memory for semantic organized material by the 21st and 35th days of treatment (p < 0. 01).
Analysis of the effectiveness of searching for numbers with switching (SCT) testified to its significant improvement: the number of errors decreased by 45 (p < 0.001) and 50% (p < 0.001) by the 21st and 35th days of treatment, respectively. At the same time, along with a decrease in the total number of errors, their quality changed - the loss of direction, the replacement of color decreased, which indicates an improvement in selectivity, greater stability of the amount of RAM.
By the 21st day of treatment, there was a decrease in the severity of autonomic disorders: headaches, dizziness, noise and ringing in the ears in 30 (75%) patients; by the 35th day of treatment, headaches and dizziness disappeared completely in 28 (70%) patients, in the remaining 12 (30%) patients they appeared extremely rarely and were slightly pronounced. Noise and ringing in the ears completely disappeared in 26 (65%) patients, in the rest the degree of its severity and frequency of occurrence significantly decreased.