POPULARITY
Host Dr. Shannon Westin and guests Dr. Bill Aronson discuss the article "High Omega-3, Low Omega-6 Diet With Fish Oil for Men With Prostate Cancer on Active Surveillance: The CAPFISH-3 Randomized Clinical Trial" and how Omega-6 are predominant in the American diet while the study significantly lowered the intake of Omega- 6 fats. TRANSCRIPT Dr. Shannon Westin: Hello everyone and welcome to another episode of JCO After Hours, the podcast where we get in depth on manuscripts published in the Journal of Clinical Oncology. I'm your host, Dr. Shannon Westin, GYN Oncologist by trade and one of the grateful Social Media Editors of the JCO. And I am very excited to welcome a special guest today, Dr. William Aronson. He is professor of Urology in the UCLA Department of Urology, the Chief of Urology at Olive View UCLA Medical Center, and Chief of Urologic Oncology at the Veterans Administration West Los Angeles. Welcome, Dr. Aronson. Dr. William Aronson: Thank you, Shannon, and delighted to be here. Dr. Shannon Westin: We are so excited to have you discussing your manuscript, “High Omega-3, Low Omega-6 Diet With Fish Oil for Men With Prostate Cancer on Active Surveillance: The CAPFISH-3 Randomized Clinical Trial,” which was published in the Journal of Clinical Oncology on December 13, 2024. So let's get right to it. First of all, you know we have a very mixed audience, so can you just level set for us and speak about the population you studied in this important trial - that low risk, favorable, intermediate risk prostate cancer. How common is that? How is it defined? That would really help. Dr. William Aronson: I would say about 50% of the patients that we diagnose with prostate cancer either have low risk disease or what we call favorable intermediate risk disease. So when the pathologists look at the cancer under the microscope, they assign what's called a Gleason grade. Grade 3 is the slower growing type of prostate cancer, grade 5 is the fastest growing type, and grade 4 is somewhere in between. So a low risk group would be only the grade 3, the slower growing type. And the favorable intermediate risk group would actually be the grade 3+4, which means they mostly see the low risk type in there, but they also see the slightly faster growing type, grade 4. So this is what we typically see. We see these patients on a very regular basis when they're newly diagnosed with prostate cancer. Dr. Shannon Westin: Okay, got it. And then can you walk us through just what the management options are typically for this patient population? Dr. William Aronson: So typically for what we call the low risk group, the patients with a low PSA and only that grade 3 type, slower growing type of prostate cancer, the standard recommendations are active surveillance. So typically, we'll periodically monitor these patients with PSA blood testing and periodically do prostate biopsies depending upon the patient's other medical problems. Dr. Shannon Westin: So I think it would also be really helpful just to understand what your typical management options are for this patient population. Dr. William Aronson: So for patients with low risk prostate cancer, they only have the Gleason Grade 3+3 with a low PSA. The standard practice is observation. And so these men will periodically see them and measure their PSA values. And periodically, they'll undergo prostate biopsy to make sure they're not getting progression of their disease. For men with favorable intermediate risk prostate cancer, that's a little different. In some practices, the patient and the urologist will decide to do active surveillance. In other scenarios, these patients will definitely elect treatment, either with radical prostatectomy or radiation therapy or other treatments that are available. Dr. Shannon Westin: So your manuscript notes that there was a high level of interest in dietary supplements and approaches among patients with prostate cancer that do elect for active surveillance. Prior to the results of CAPFISH-3, did we have any data to support those types of recommendations? Dr. William Aronson: We actually don't have any long term prospective randomized trials that support that recommendation. There have been a number of very interesting epidemiologic studies, for example, suggesting maybe a plant-based diet might be helpful. Or a number of other studies suggesting maybe more tomato-based products like tomato sauces or tomato paste may be helpful. But no prospective longer term randomized trials that were positive. Dr. Shannon Westin: Okay, that makes sense. So what led you all to explore the high omega-3, low omega-6 fatty acid diet in this trial? Dr. William Aronson: After our initial omega-6 studies, we subsequently did some studies where we raised the omega-3 from fish oil and lowered the 6, looking at a more favorable ratio of the omega-3 to omega-6. And once again, we found that in our animal models, there was a significant delay in progression of prostate cancer. That then led us to perform a clinical trial. It was a short term trial in men prior to undergoing radical prostatectomy. And in these men, they were randomly assigned to one of two groups, either a western high fat diet or a low fat diet with fish oil. And we found after just four to six weeks, a significant change in the Ki67 level in their radical prostatectomy tissue. And Ki67 is actually a strong indicator of prostate cancer progression, spread, or even death from prostate cancer. Dr. Shannon Westin: Well, and I think that leads us really nicely into the design of the current study. So why don't you walk us through how CAPFISH-3 was designed. And you've already spoken a little bit about your primary endpoint. Dr. William Aronson: Based on the results of what we saw in the lab and what we saw in our short term clinical trial, we decided to perform a one year trial, a longer term trial in men on active surveillance. And these men were randomly assigned to either a diet with slight reduction in dietary fat, specifically reduction in the omega-6 intake as well as increase in foods with omega-3 and fish oil capsules. The other group, we asked the men to just not take fish oil capsules, but they could eat whatever else they wanted during the course of the study. Men in the diet where we lowered the omega-6 and raised the omega-3, they were seen by a dietitian once a month to really ensure that they were compliant with that intervention, which they were. The other intriguing part of our study, which I think is super important, is the precision that was used when these men underwent prostate biopsy. So, at baseline and at one year, when these men underwent prostate biopsy, they had the same site within the prostate biopsied. That's important because it's not so easy to find the same site within the prostate because of heterogeneity throughout the prostate. And so we were able to obtain that high level of precision as they were in an active surveillance program at UCLA with Dr. Leonard Marks. Dr. Shannon Westin: So we spoke a little bit about what's important about the Ki67 index as your primary endpoint. Can you talk a little bit about what the study found with your intervention? Dr. William Aronson: So we found that the Ki67 index increased by 24% in the control group and decreased by 15% in the low omega-6, high omega-3 group with fish oil capsules. So that ended up resulting in a statistically significant change between the groups favoring the low omega-6, high omega-3 group. Dr. Shannon Westin: And then what were the secondary endpoints that CAPFISH-3 explored? Anything of note that you want to review for the listeners? Dr. William Aronson: So a number of positive secondary endpoints from the trial. Firstly, we saw that the triglyceride levels were lower, which is what can typically be seen with omega-3 intake. We also saw reduced levels of a cytokine, a circulating factor in the bloodstream called ‘macrophage colony stimulating factor'. And that's particularly interesting because there's a certain type of macrophage which is well known to be involved in prostate cancer progression in men with more advanced prostate cancer, and we've been able to inhibit that in our animal models and in our tissue culture studies. And it was especially interesting to see that we did have an effect on this particular cytokine in this prospective randomized trial. We did not see changes in a number of other measures, including Gleason grade or PSA. These are measures that we use in clinical practice. To see an effect on those would have required a longer term and larger study to be performed. Dr. Shannon Westin: That makes sense. I think it's always great to try to get as much of these types of translational data as we can. But sometimes you just have to do what is reasonable and you get what you get. It looks to me like this regimen was fairly well tolerated. Did you obtain any patient reported outcomes or feedback on the trial? Dr. William Aronson: So, there were four patients in the fish oil group that did have some side effects, and we withdrew them from the study. They did have some effects on their upset stomach, and a number of men also had some diarrhea as well. And so for those four patients, we did withdraw them from the study. Dr. Shannon Westin: And then I guess the last question I have is really, what's next for this intervention? Are we ready to move this to the clinic or what do you see as next steps? Dr. William Aronson: Well, this next step that we're working on right now is to better understand exactly what happened in these patients. So we have blood, we have tissue, we're doing genetic studies on these patients. So that's really the first step, in our mind, to better understand what happened before moving to the next step. I'm particularly intrigued about trying this intervention in men with more advanced prostate cancer, specifically because of what we see, this particular diet and how it's affecting the patient's immune system and how that may favorably affect their course of their prostate cancer. Dr. Shannon Westin: Well, great. Well, thank you so much for taking the time to chat with us about such an important clinical trial, and I really appreciate all the work you're doing and hope to get to see you soon. Dr. William Aronson: Well, thanks for having me, Shannon. It's really an exciting finding and I think it's something that clinicians and patients are going to be super interested in. Dr. Shannon Westin: We love straightforward interventions that actually make a difference, so you guys are to be congratulated for that. And I just want to thank all of you for listening. Thanks again, and I hope you enjoyed this episode of JCO After Hours. Be sure to check out our other podcast offerings wherever you get your podcasts. Have an awesome day. The purpose of this podcast is to educate and to inform. This is not a substitute for professional medical care and is not intended for use in the diagnosis or treatment of individual conditions. Guests on this podcast express their own opinions, experience, and conclusions. Guest statements on the podcast do not express the opinions of ASCO. The mention of any product, service, organization, activity, or therapy should not be construed as an ASCO endorsement. Dr. Aronson Disclosures: Stock and Other Ownership Interests Johnson and Johnson Speakers' Bureau Company name: Janssen Oncology, Bayer, Blue Earth Diagnostics, AstraZeneca, Pfizer/Astellas Research Funding: Lantheus Medical Imaging UCLA Health Article Video
Send us a textWelcome back to the DigiPath Digest, fresh from PathVision! In this episode we will dive into the latest updates from the PathVision conference, covering trends in AI-driven diagnostics, the expansion of digital pathology into primary care, and the exciting new frontier of glassless pathology.Join me as I recap the highlights of PathVision and the latest updates from the digital pathology literature, including discussions on:AI Integration in Pathology: Learn how AI is advancing breast cancer diagnostics with tools like Ki-67 scoring models and multi-label AI for mammography, aimed at reducing unnecessary biopsies.Global Health & Digital Microscopy: Hear about innovative projects from Sweden and Finland focused on AI-supported digital microscopy in primary healthcare labs, bringing accessible diagnostics to underserved areas.Glassless Pathology with MUSE: Discover how glassless pathology is changing tissue imaging with MUSE (Microscopy with UV Surface Excitation), enabling diagnostics without the need for traditional glass slides. Dr. Zuraw breaks down what this means for future pathology workflows.Plus, a shout-out to the vendors and partners making these advancements possible, and insights from Dr. Zuraw's conversations with digital pathology trailblazers from around the globe, including new developments from Asia in digital pathology education and technology.Timestamps:[0:00] PathVision Highlights & Global Attendees[5:15] AI in Diagnostic Workflows: Dr. Anil Parwani's “Pathology Train Ride”[12:30] Moving Beyond Narrow AI: Multimodal and Foundational Models[18:45] Glassless Pathology: A New Frontier with MUSE Microscopy[25:10] Integrating Digital Microscopy in Global Health Labs[32:00] Breast Cancer Month: New Advances in AI for Diagnostics[42:00] One Health & AI for Disease Detection in Primary Care[48:30] Special Interviews: Jun Fukuoka and Asian Society of Digital PathologyLinks and Resources:Subscribe to Digital Pathology Podcast on YouTubePathology News Signify Research Monthly RecapYouTube version of this episodePublications Discussed Today:
Das Ziel der Gebärmutterhalskrebsvorsorge ist es, auffällige Zellen und Krebsvorstufen rechtzeitig und verlässlich zu erkennen. Werden beim Pap- oder dem HPV-Test Auffälligkeiten festgestellt, sind unter Umständen weitere Untersuchungen notwendig, um abzuklären, ob es sich tatsächlich um eine Krebserkrankung handelt. Welche Bedeutung Biomarker wie p16/Ki67 für die Früherkennung von Zervixkarzinomen und entsprechende Vorstufen haben, erklärt unser heutiger Gesprächspartner Dr. Gerd Böhmer.
Host: Mary Katherine Cheeley, PharmD, BCPS, CLS, FNLA Guest: Leigh Anne Cantrell, MD, MSPH The updated ASCCP guidelines now address the use of dual-stain technology for cervical cancer management. CINtec PLUS Cytology is a test for the co-expression of p16 and Ki67 and confirms the presence of a transforming HPV infection and defines patients with high-risk cervical disease. To learn more about these guidelines and how to apply them in practice, join Dr. Leigh Cantrell, Medical Physician and Fellow in the American College of Obstetrics and Gynecology. COBAS and CINTEC are trademarks of Roche.All other product names and trademarks are the property of their respective owners. 07/24
Host: Mary Katherine Cheeley, PharmD, BCPS, CLS, FNLA Guest: Leigh Anne Cantrell, MD, MSPH The updated ASCCP guidelines now address the use of dual-stain technology for cervical cancer management. CINtec PLUS Cytology is a test for the co-expression of p16 and Ki67 and confirms the presence of a transforming HPV infection and defines patients with high-risk cervical disease. To learn more about these guidelines and how to apply them in practice, join Dr. Leigh Cantrell, Medical Physician and Fellow in the American College of Obstetrics and Gynecology. COBAS and CINTEC are trademarks of Roche. All other product names and trademarks are the property of their respective owners. 07/24
Host: Mary Katherine Cheeley, PharmD, BCPS, CLS, FNLA Guest: Leigh Anne Cantrell, MD, MSPH The updated ASCCP guidelines now address the use of dual-stain technology for cervical cancer management. CINtec PLUS Cytology is a test for the co-expression of p16 and Ki67 and confirms the presence of a transforming HPV infection and defines patients with high-risk cervical disease. To learn more about these guidelines and how to apply them in practice, join Dr. Leigh Cantrell, Medical Physician and Fellow in the American College of Obstetrics and Gynecology. COBAS and CINTEC are trademarks of Roche.All other product names and trademarks are the property of their respective owners. 07/24
CME credits: 0.25 Valid until: 29-05-2025 Claim your CME credit at https://reachmd.com/programs/cme/consensus-recommendations-for-use-of-p16ki67-dual-staining-cytology-in-the-management-of-individuals-testing-positive-for-hpv/14675/ More effective triage tests are essential to improving cervical cancer risk-stratification and thus reducing the potential for unnecessary colposcopies. Enduring consensus cervical cancer screening recommendations, published in 2024, describe specific-use algorithms for p16/Ki-67 dual-staining cytology as an effective triage test, and also offer insight on how this test can best fit into clinical practice. Join Drs. Warner Huh and Kimberly Levinson as they first establish the rationale for using p16/Ki-67 dual-staining cytology and then break down the most salient algorithms for its use as offered in the recent recommendations.=
BUFFALO, NY- December 12, 2023 – A new #researchpaper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 15, Issue 22, entitled, “Tat-heat shock protein 10 ameliorates age-related phenotypes by facilitating neuronal plasticity and reducing age-related genes in the hippocampus.” In this new study, researchers Hyo Young Jung, Hyun Jung Kwon, Kyu Ri Hahn, Woosuk Kim, Dae Young Yoo, Yeo Sung Yoon, Dae Won Kim, and In Koo Hwang from Seoul National University, Chungnam National University, Gangneung-Wonju National University, Hallym University, and Konkuk University investigated the effects of heat shock protein 10 (HSP10) protein on memory function, hippocampal neurogenesis, and other related genes/proteins in adult and aged mice. “In the present study, we investigated the effects of HSP10 on hippocampal function in both adult and aged mice.” To translocate the HSP10 protein into the hippocampus, the Tat-HSP10 fusion protein was synthesized, and Tat-HSP10, not HSP10, was successfully delivered into the hippocampus based on immunohistochemistry and western blotting. Tat-HSP10 (0.5 or 2.0 mg/kg) or HSP10 (control protein, 2.0 mg/kg) was administered daily to 3- and 21-month-old mice for 3 months, and observed the senescence maker P16 was significantly increased in aged mice and the treatment with Tat-HSP10 significantly decreased P16 expression in the hippocampus of aged mice. In novel object recognition and Morris water maze tests, aged mice demonstrated decreases in exploratory preferences, exploration time, distance moved, number of object contacts, and escape latency compared to adult mice. Treatment with Tat-HSP10 significantly improved exploratory preferences, the number of object contacts, and the time spent swimming in the target quadrant in aged mice but not adults. Administration of Tat-HSP10 increased the number of proliferating cells and differentiated neuroblasts in the dentate gyrus of adult and aged mice compared to controls, as determined by immunohistochemical staining for Ki67 and doublecortin, respectively. Additionally, Tat-HSP10 treatment significantly mitigated the reduction in sirtuin 1 mRNA level, N-methyl-D-aspartate receptor 1, and postsynaptic density 95 protein levels in the hippocampus of aged mice. In contrast, Tat-HSP10 treatment significantly increased sirtuin 3 protein levels in both adult and aged mouse hippocampus. These suggest that Tat-HSP10 can potentially reduce hippocampus-related aging phenotypes. “Our results suggest that Tat-HSP10 treatment facilitates mitochondrial function, and Tat-HSP10 supplementation ameliorates the aging phenotypes in the mouse hippocampus.” DOI - https://doi.org/10.18632/aging.205182 Corresponding authors - Dae Won Kim - kimdw@gwnu.ac.kr, and In Koo Hwang - vetmed2@snu.ac.kr Keywords - aging, heat shock protein 10, hippocampus, memory, neurogenesis, synaptic plasticity About Aging-US Launched in 2009, Aging-US publishes papers of general interest and biological significance in all fields of aging research and age-related diseases, including cancer—and now, with a special focus on COVID-19 vulnerability as an age-dependent syndrome. Topics in Aging-US go beyond traditional gerontology, including, but not limited to, cellular and molecular biology, human age-related diseases, pathology in model organisms, signal transduction pathways (e.g., p53, sirtuins, and PI-3K/AKT/mTOR, among others), and approaches to modulating these signaling pathways. Please visit our website at https://www.Aging-US.com and connect with us: SoundCloud - https://soundcloud.com/Aging-Us Facebook - https://www.facebook.com/AgingUS/ X - https://twitter.com/AgingJrnl Instagram - https://www.instagram.com/agingjrnl/ YouTube - https://www.youtube.com/@AgingJournal LinkedIn - https://www.linkedin.com/company/aging/ Pinterest - https://www.pinterest.com/AgingUS/ Media Contact 18009220957 MEDIA@IMPACTJOURNALS.COM
Drs. Hope Rugo and Nancy Davidson discuss the next generation of endocrine therapy in hormone receptor-positive breast cancer – SERDs, SERMs, CERANs, and PROTACs – and challenges relating to the optimal sequence of therapeutic options. TRANSCRIPT Dr. Hope Rugo: Hello, I'm Dr. Hope Rugo, your guest host of the ASCO Daily News Podcast today. I'm a professor of medicine and director of breast oncology and clinical trials education at the University of California San Francisco's Comprehensive Cancer Center. Hormone receptor-positive (HR+) breast cancer is the most common subset of this disease and breast cancer is the most common cancer diagnosed in women worldwide. Endocrine therapy (ET) is the cornerstone of management in hormone receptor-positive breast cancer and may involve suppressing estrogen production with aromatase inhibitors in the cancer cell itself, or directly blocking the estrogen receptor pathway through selective estrogen receptor modulators, such as tamoxifen, or the injectable selective estrogen receptor degrader, fulvestrant. However, despite the availability of these therapies, the largest unmet need lies in treatment of HR-positive HER-negative disease lies after progression on endocrine therapy. On today's episode, we'll be discussing the emerging generation of endocrine therapies in breast cancer, some of which were designed to overcome common mechanisms of endocrine resistance, and the challenges relating to the optimal sequence of therapeutic options. Today, I'm delighted to welcome the world-renowned breast cancer researcher, Dr. Nancy Davidson, to the podcast. She's a professor and the executive vice president for clinical affairs at the Fred Hutchinson Cancer Center and professor of medicine at the University of Washington as well as past president of ASCO and AACR. You'll find our full disclosures in the transcript of this episode and disclosures of all guests on the podcast can be found in our transcripts at asco.org/DNpod. Nancy, it's great to have you on the podcast today. Thanks so much for being here. Dr. Nancy Davidson: Thank you so much, Hope, for the opportunity. Dr. Hope Rugo: You've done incredible work in the area of treating HR+ disease, but also in understanding some of the agents and pathways that are most important to our understanding of how we treat and really think about developing new drugs for HR+ breast cancer. The first generation of anti-estrogen drugs really included selective estrogen receptor modulators, but now we have a new generation of these anti-estrogen drugs. And there's also orally administered selective estrogen receptor degraders, so-called SERDs, that we hope someday will be an alternative with already one approved for the injectable SERD, fulvestrant. But there's a whole additional class of drugs, complete estrogen receptor antagonists, referred to as CERANs, and proteolysis targeting chimerics, PROTACs. These agents are all at various stages of development in both early and metastatic settings and really represent, I think, a confusing array of different treatments that are being studied, some of which of course are approved. Nancy, it would be great if you could tell us some about these exciting new agents, starting with our approved drugs and moving on to the agents that we're studying in clinical trials. Dr. Nancy Davidson: Hope, this is a terrific example of how basic science has really begun to inform clinical practice. For us as breast cancer practitioners, it was really easy for a long time because we only had tamoxifen. And then things like the aromatase inhibitors as a means of decreasing the estrogen ligand came into practice, as did fulvestrant, as you mentioned, a selective estrogen receptor degrader. Thanks though to a lot of understanding about estrogen receptor biology, and specifically, the discovery about a decade ago of estrogen receptor mutations, something that we didn't think exist ed for a long time, but which we now know exists in maybe a third to a half of breast cancers after exposure to aromatase inhibitors, that's led to really an explosion, this alphabet soup of possibilities that you talked about. SERMs, selective estrogen receptor modulators, tamoxifen is the classic one—something that acts as an agonist and as an antagonist, depending on the tissue. The SERDs, the selective estrogen receptor degraders, lead to estrogen receptor destruction. Fulvestrant is the classic example of this right now. But we're very excited that a second one has been FDA approved. That's elacestrant that was approved earlier this year, and it has specifically been developed to try to target those estrogen receptor mutant breast cancers because those are the ones that are particularly resistant. So that's the first of this new generation of SERDs that's coming on the market, has come on the market. As you say, a key advantage there is that it's oral, unlike fulvestrant, which requires 2 monthly injections and is very, very inconvenient and uncomfortable for patients. Other SERDs are coming along. I think another one that is well along in clinical development is giredestrant, which is in the same family and being tested in clinical trials of the classic varieties. It's been tested in a window trial, looking at its ability to down-regulate Ki67. It's being tested against standard of care in first-line metastatic breast cancer. And there are some studies that are going to begin looking at it in the adjuvant setting as well. There are other members of this particular family, but I think that elacestrant is the one that we have at our fingertips right now, and giredestrant is one that's certainly coming along in clinical trials, and we should look forward to those results. For those who are interested in the giredestrant, the trials in question right now that are going on are persevERA, which is in stage 4 breast cancer, and lidERA, which is in the adjuvant setting. Now that's one new category, or it's an old category with a new twist. A second is these agents that are called complete estrogen receptor antagonists, CERANs, as you talked about them. The key here is that what they do is, the estrogen receptor has a couple of domains, and in particular, it has 2 different activation domains, AFT1 and AFT2. And these CERANs are complete estrogen receptor antagonists, so they block both of those domains. And the hope would be that they might end up being more effective than the other agents that we have available to us right now. So those are also in clinical trial at the present time, and we're waiting to see whether or not there's going be any value in that particular area that's going to able to go forward for us. Another area that I think we want to talk about is the PROTACs, as you mentioned. These are proteolysis targeting chimeric entities. So, this is a kind of a complicated situation where you have a kind of bivalent situation where you have something that binds both to the estrogen receptor and then also to E3 ubiquinase. So, it leads to degradation again of the agents; that's another area for us to be watching. And then finally the SERCAs. So those are the selective estrogen receptor covalent antagonists, and what they do is they bind very specifically to a cysteine 530 that exists in the estrogen receptor but not in other steroid receptors. So that's where the selective part of this comes along. Now these are all in various stages of clinical investigation. Some of them are pretty early at this point, but I think the ones that are well established are elacestrant, giredestrant is coming along as we just talked about. Camizestrant is also coming along through the SERENA series of trials, and imlunestrant is also coming on through the EMBER series of trials. Hope, I would also be remiss if I didn't go back to an old drug that's a new drug, and that's lasofoxifene, which is also in the SERM family. Those who have been in our field for a while will remember that lasofoxifene was actually originally kind of tested in the prevention setting, where it seemed to have some activity. It came back into our interest because it has really strong activity against estrogen receptor mutant breast cancer models in the laboratory. And as a consequence of that, it's coming back into the clinic through a series of trials that are called the ELAINE trials, where we're looking to see whether or not it might also be better than fulvestrant. It too is an oral agent, so that's a real plus for us, and the first set of ELAINE trials would suggest that there's some nice activity without a lot of toxicity. So, lots going on in this field. You know, I think for all of these things, obviously, we're also working very hard to think about the toxicities. All of these, as I recall, are oral agents. So that gets rid of 1 huge toxicity, which is you don't have the need for some sort of injection. But they all do have some side effects. Frequently, [the toxicities] are like GI side effects or fatigue. In a couple of cases, there might be some concern about cardiac arrhythmias, but I think GI turns out to be one of the most important things that we have seen in some of these trials. Generally, it's very well tolerated, though. I think another important question, which I'm sure is in your head and is in mine, is that how are we going to integrate these into what we already have? And so, I think a lot of the work right now is looking at patients who have already received an aromatase inhibitor plus a CDK4/6 inhibitor and are now going on to one of these new agents. But you might wonder, if they turn out to be effective and well-tolerated, whether they too should be perhaps combined with CDK4/6 inhibitors in place of the aromatase inhibitors or the fulvestrant that we use now. So, I think that we can imagine that those clinical trials are either in progress in some cases or will be coming on as we try to think about how to integrate these new approaches into our existing standard of care, which is already quite complicated, right? We've gone far from tamoxifen, which was good for everybody, to now a really complex algorithm about how we think about hormone therapy, both in early breast cancer and in metastatic breast cancer. Dr. Hope Rugo: Well, that was a fabulous discussion about the new agents and our existing agents and both the exciting aspects, as well as the challenges. One question that comes up, I think a lot, and this is of course a huge question in many ways, but for ER-positive metastatic breast cancer, where these drugs are first being tested, maybe we'll talk about that first. There's one trial at SERENA-6, looking at camizestrant in patients who have developed evidence of an ESR1 mutation in circulating tumor DNA, who are on first-line therapy with an AI and a CDK4/6 inhibitor. What do you think about that study and where do you think that progress might go? Of course, that's based on data from another trial, which wasn't definitive, but suggested, had sort of a suggestion, you know, without studying the sequencing in detail that maybe you would have improved progression-free survival with that approach. Dr. Nancy Davidson: Yeah, by which you mean the idea of monitoring pre-ESR1 mutations and circulating DNA and then making therapy changes based on that? Is that what you're talking about? Dr. Hope Rugo: The PADA-1 trial, yes, and that led to this huge SERENA-6 trial, which of course is now accruing. Dr. Nancy Davidson: So, you know, we would love that, right? I think that obviously breast cancer for many years has been interested in trying to have circulating markers that we could use to help to guide our therapy in a more meaningful way than we have in the past. And I think that the trial that you've talked about is certainly one that's trying to make that see whether that's a possibility for us based on information like the PADA trial. I'm hopeful it's going to work out. I would say, let's see what that looks like, whether it's going to be useful or not. It seems to me in some cases, some of these trials have not been quite what we hoped for, but I don't think we necessarily had the molecular techniques or the sensitivity, nor did we necessarily have other things to move to because, you know, those two approaches require both a really good test, but also the ability to use the test to define a new therapy that's going to lead to improved patient outcomes. And I think that these are ingredients that we now have available to us at our fingertips more than we did, say, a decade or 2 decades ago. Exciting approach. Dr. Hope Rugo: I do think there's a lot of challenges inherent in this process, monitoring blood on a regular basis and following up and then randomizing based on results. But I think [SERENA-6] is an incredibly important trial, as you have mentioned, to try and think about moving past using circulating tumor cells, which didn't work, to just change blindly to the next therapy, but more have a rational reason to change to a drug that may be more effective before the disease itself progresses. And just for our listeners, the unique aspect of trials like this in SERENA-6 is that you change therapy before you have evidence of disease progression, but only this molecular evidence of a mutation that is associated with resistance to the therapy you're on. So, it's a really important question. We'll see what happens, as you mentioned, sometimes we're not clever enough to really get around the fact that there are multiple mutations driving resistance in this setting, so we'll see how straightforward this is. The question also comes up, and I think that's a question with many of these trials now, is they're randomizing patients after a progression on an AI and CDK4/6 inhibitor to receive the novel endocrine therapy or fulvestrant. One of, I think, the concerns of treating oncologists is that then you're sort of eliminating the possibility of a targeted agent in that setting. And of course, we have new targeted agents we're studying as well, AKT inhibitors and new inhibitors of the PI3 kinase pathway. So, the newer trials are now combining, as you mentioned. There's a lot of concerns about drug-drug interactions here and how you might really combine them. And then there are triplet studies looking at CDK4/6 inhibitors, oral SERDs primarily, and pi3-kinase and CDK4/6 inhibitors. What's your thought on these and will they really help to move the needle forward? Dr. Nancy Davidson: I think that's a really interesting question, Hope, is whether or not we're going to find that combination therapies from the get-go are the way to go, or whether we're going to end up having to use maybe more serial therapies. Because not only is it a question about whether or not you can tackle all of these different resistance mechanisms simultaneously, but I think the other question is, as you say, are there negative drug-drug interactions, and are there toxicities that are intolerable? Although these are targeted in all cases and they're relatively benign in terms of side effects from the breast cancer perspective, they're not devoid of toxicity. And so, I think that's going to be another issue for us – whether they're well tolerated during the time that patients are taking them. I guess the other thing I always think about, Hope, is that it's hard to know about value of cancer care, but you know, we are talking about agents that are not inexpensive. You know, when you think about the financial toxicity, in addition to the side effect toxicity that you and I just talked about, trying to think about what that value is going to look like is going to be very important for us. Also, as somebody who worked for a long time in the lab and in the clinic, you know, there are an infinite number of combinations that you and I could think about that are rational. And so, the question is, how are we going to pick the ones that are the most rational, if you will, the ones that seem to be the most promising, and take them forward into clinical trials? Because patients are our most precious resource. And so, we want to make sure that we are bringing forward only those things that really seem to have a very strong foundation and the opportunity to improve outcomes over time. Tough, tough question for us to try to think about, as you've talked about. The other thing is that, you know, these trials are not only for postmenopausal women, who are the majority of patients, but we also want to target them to premenopausal women. So, in those women, we're also looking at using an LHRH agonist on top of this, right, because many of these things are really at least so far designed for the postmenopausal state. So stay tuned. Lots of work going on. I think one of the interesting things will be making the leap from using these in metastatic disease at time of progression to taking them forward into the early breast cancer space. And several of the agents are now beginning to do that because of very strong preclinical and clinical data to date. Dr. Hope Rugo: So, we have new agents that we're studying in the metastatic setting, and we've seen a trend to move fairly rapidly from phase 1B trials directly into phase 3 trials, because as you pointed out so clearly, there are a number of drugs in this setting, and we don't really know not only which agent is better, but even what class of agents is better. So, as we move more quickly from phase 1B to phase 3, the question comes up about how we're going to study these agents optimally in the early-stage setting. They, I think we all thought that maybe changing based on ctDNA evidence of a mutation might be an approach, but that's complicated. A big question for you is whether or not you think that's ready for prime time as it's being marketed as such. And then the second question is really, are we better changing our whole approach upfront in high-risk disease or should we wait until after patients are exposed to their endocrine therapy and then switch as we go along; the EMBER-4 trial is looking at that switching approach? Dr. Nancy Davidson: Yeah, I think that this is obviously the billion-dollar question for many companies and for many of us as investigators. I don't know that I have a crystal ball into what the best approach might be. We know that already some of these have been abandoned. For example, amcenestrant. So, I don't even have to learn how to say it because it has been abandoned. It did go to phase 3, as you pointed out, in the advanced setting, looking at it with palbo vs letrozole plus palbo, and it didn't really show a whole lot that the company wanted to pursue. So, I think that sometimes these things are going to be abandoned based on the metastatic setting, which is a large trial, and that's a trial where obviously it might be a little easier to the circulating markers, as you talked about, than maybe in the early-stage breast cancer setting. I think that probably these early-stage trials are going to end up being big; they're going to have be clean. I'm guessing that most companies and most investigators will want to target them towards high-risk individuals, as you talked about, for 2 reasons. One is that ethically, I think we feel more comfortable with that. These are individuals where standard therapies are maybe not serving them as well as we would like and where we have information to think that we can at least have equipoise about a new approach. And the second is that from a trial design point of view, the event rate is likely to be higher, and therefore you might get answers earlier and with a smaller clinical trial. So that's where I suspect we're going to go. But it's a challenging question, and I think that many investigators and many companies are really trying to struggle with that right now because we do have so many options. And we're not quite sure how to, first, develop these new drugs, but then really importantly, put them in the context of our existing drugs. And as you say, we're also simultaneously trying to develop superior molecular or circulating markers to guide us. So, there's so many variables that are going on right now in this field that, from my point of view, itmakes it really exciting, but it also makes it pretty complicated, both for investigators and for pharma as we try to think about how to position these going forward. But what a great problem to have, Hope, because remember when you and I started in breast cancer, pretty much all you needed to know was tamoxifen. And I think you and I also probably started at the tail end of the time when we were using androgens and progestins and agents that now have basically completely fallen out of favor. Dr. Hope Rugo: Estrogen Dr. Nancy Davidson: Estrogen, yeah, and your side effect profiles were not as good. So, it's a good problem to have. It's a nice situation when science can inform our clinical investigation, our clinical practice. Dr. Hope Rugo: Yes, I think it'll be fascinating to see where we end up at the end, but of course, the complexities of this include the fact that sometimes just the clinical trial design leads to a less than positive result because of the way the trial itself has been designed, which I think is the nice part about moving these earlier into treatment of high-risk disease, but also brings up the question of all sorts of areas we're not really a place to discuss today, like how the statistical design is made, et cetera, that can sometimes result in poor evaluation of excellent agents. One of the questions that comes up, and you brought this up earlier, which I think is really important, is that we are in the era of combining our endocrine agents with targeted agents, and, oral agents, in a fascinating way, really bring up drug-drug interactions in a way that the injectable fulvestrant hasn't, and aromatase inhibitors were kind of quiet on the impacting metabolism, unlike tamoxifen. But we are seeing some drug-drug interactions and certainly the discussions about using these agents may include the question about whether or not you're having more diarrhea or nausea or fatigue or one drug causes photopsia, flashing lights, things like that. Keratitis is becoming a new toxicity to follow. How are we going to figure out how to sequence these drugs and are they only going to work better all of them as a class in patients with ESR1 mutations in their tumors? Dr. Nancy Davidson: I think we don't know the answer to the second question yet. That's something that really needs to be sorted out. Even if they do, that's still a really important subset of patients, assuming that we continue to start with the aromatase inhibitors, right? That's where those things really seem to emerge right now. I don't know how we're going to figure those things out. I guess that I'm hoping that maybe a couple of agents in these classes will become kind of the lead agents. And so, we'll be able to do this work with a handful of things as opposed to a whole array of things. But we'll see. I think that even within classes, obviously, these agents are going be slightly different, probably. And so it may be that one member of a class may be slightly better from a, maybe not from an efficacy point of view, but from a toxicity point of view. And we'll just have to see how it goes. I don't think I have any magic answers about that. Dr. Hope Rugo: Yes, it'll be interesting to see whether or not the agents work in sequence too. You know, could you use a PROTAC after an oral SERD, for example, or a CERAN? That'll be fascinating to see. Dr. Nancy Davidson: I'm hoping that preclinical modeling may help with that a little bit, though of course we all know that there are plenty of things that do well in preclinical models, GEMS and rodent models and PDXs, and sometimes those things translate nicely into clinical practice, but sometimes they don't. Dr. Hope Rugo: You mentioned, Nancy, that when we started out, that it was a fairly simple decision about what endocrine therapies, and we ran out very quickly. We're seeing some of those old classes come up with new agents. And you mentioned lasofoxifene, the oral SERM that seems to have some benefits and works in the later-line setting and also can be combined with a CDK4/6 inhibitor; [and also] has data with abemaciclib. There's also a new androgen receptor agonist, enobosarm, that's being tested as well. Do you think that these older mechanisms have a future as well? Dr. Nancy Davidson: I do. I think that because we'll be able to understand the biology better than we did in the past. A lot of our hormone therapy was pretty empiric several decades ago. But I think with better understanding of mechanisms and better understanding of what the patterns of resistance might be for a particular tumor, that we might be able to think about those things. You're right that there's a whole parallel universe right now in androgen receptor targeted therapies that we're not talking about today, both in perhaps in hormone-responsive breast cancers, but also in triple-negative breast cancers in a certain subset. And so that's an area where we probably need to be watching what our prostate cancer colleagues are doing as they develop these agents and thinking about where they might mechanistically make sense to apply in the breast cancer field. It won't be the first time that we've received insights from them because remember, prostate cancer people knew about androgen receptor mutations a really long time before we figured out that they existed in estrogen receptor as well. So, there's a nice cross talk, I think, between the prostate cancer field and the breast cancer field in that regard. Dr. Hope Rugo: That's an excellent point. And we learn a lot from our colleagues studying other malignancies, and prostate cancer has been a big area there for us and hopefully will help us with studying these agents because there's different toxicity profiles, of course, as well. And then you mentioned the approval of elacestrant, the first oral SERD to have regulatory approval, and it's approved in patients who have ESR1 mutations in their tumors. We're also studying it in the ELEVATE trial in combination with all of the different targeted agents, the CDK4/6 inhibitors, mTOR inhibitor, everolimus, and the PI3 kinase inhibitor, alpelisib, to really try and understand how these can be optimally combined. Would you check for ESR1 mutations at diagnosis of metastatic disease, or would you do this more after progression or start of progression of disease on an aromatase inhibitor? Dr. Nancy Davidson: I tend to be a conservative, Hope, and so unless there's a clinical trial that might be able to be considered, I would tend to check it only after progression on the aromatase inhibitor. But I think some people are earlier adopters, and I suppose it's possible that they might want to know from the get-go. Having said that, I do think elacestrant has an approval after aromatase inhibitor, as I recall. So, presumably the patient would have to have that exposure before you would be able to act on the ESR1 mutation by administering the oral SERD. This is also a new area that may well change with time, right? You know, as we develop different agents, as the agents have different requirements or different indications, and as we perhaps have better tests, it may be things that we don't do routinely now will become very routine in the future. And perhaps in series like you talked about in a serial fashion. Dr. Hope Rugo: Absolutely. I think that's a really important comment about how we're going to think about treating hormone receptor positive disease and potentially, you know, we have new chemo options, of course, not part of our talk discussion today and antibody drug conjugates. So, the future is certainly challenging in terms of understanding this appropriate sequencing. We need data, but it's exciting to have these options. As you pointed out, this is this is a great problem to have, you know, too many drugs with efficacy to try and understand how to use these in the most appropriate way. And as you also pointed out, the use in young women is particularly important. Whether or not you need to suppress the ovaries with all of these new agents is going to be important to understand as well as a next step. I don't know if you have specific thoughts on that area as well. I mean, that may reduce toxicity for younger women. Dr. Nancy Davidson: I guess our approach right now has been to suppress them, but I agree with you; you wonder if you look at some of the mechanisms of action, whether that's really a requirement biologically and medically, or whether it's something that's kind of a leftover from the approval process. You know, that for the drug approval, these women were suppressed. I think we need to work that out because that's another area where certainly women could have ovaries removed. That's pretty straightforward, although it requires a surgical procedure. But otherwise use of LHRH agonists continues to be injections, right? And so, if we're trying to minimize convenience or maximize convenience and minimize toxicity for women with any stage of breast cancer, anything we can do, it seems to me to eliminate an injection is going to be a good thing. So important for us to be able to work that out as well, even though numerically it's a smaller number of women, it's obviously a very important number. population of women. Dr. Hope Rugo: Last question for you. One question that comes up and sent to me and panels all the time is, if you have done your next generation sequencing and you see an ESR1 mutation and a P13CA mutation, what do you prioritize in terms of your choice of treatment, because we have the choice of either using elacestrant or a fulvestrant in combination with an agent targeting the PI3 kinase pathway? Dr. Nancy Davidson: I don't know that there's a right answer to that medically right now. So, when I talk about it with patients, I suggest to them that it's probably not one or the other. It's kind of the question of which one to use first. So, we talk a little bit about side effect profiles. We talk a little bit about patient preference and neither of these drugs is devoid of problems, but sometimes patients have pretty strong feelings about which set of side effects seems the least unattractive to them or the possibility of having those side effects. And I know that if one isn't well tolerated, you can swap over to the other. Dr. Hope Rugo: Absolutely. I think that's a very important way to think about next therapies in our era of not really knowing what's right. And this idea of shared decision-making is so incredibly important. Nancy, thank you so much for sharing your insights and knowledge with us today. I could talk to you for hours about this. Dr. Nancy Davidson: Hope, thank you so much for the opportunity. It is an exciting area and I really enjoyed talking with you about what we know and the many things that we don't yet know, but we're working on. Dr. Hope Rugo: Indeed, it's certainly an exciting time. Thank you to our listeners for joining us today. And thanks to the ASCO Daily News Podcast for highlighting this important area. You'll find a link to all of the studies discussed today in the transcript of this episode. Finally, if you value the insights that you hear on the ASCO Daily News Podcast, please take a moment to rate, review, and subscribe wherever you get your podcasts. Thanks so much. Disclaimer: The purpose of this podcast is to educate and to inform. This is not a substitute for professional medical care and is not intended for use in the diagnosis or treatment of individual conditions. Guests on this podcast express their own opinions, experience, and conclusions. Guest statements on the podcast do not express the opinions of ASCO. The mention of any product, service, organization, activity, or therapy should not be construed as an ASCO endorsement. Follow today's speakers: Dr. Hope Rugo @hoperugo Dr. Nancy Davidson Follow ASCO on social media: @ASCO on Twitter ASCO on Facebook ASCO on LinkedIn Disclosures: Dr. Hope Rugo: Consulting or Advisory Role: Napo Pharmaceuticals, Puma Biotechnology, Mylan, Eisai, Daiichi Sankyo Research Funding (Inst.): OBI Pharma, Pfizer, Novartis, Lilly, Genentech, Merck, Daiichi Sankyo, Sermonix Pharmaceuticals, AstraZeneca, Gilead Sciences, Astellas Pharma, Talho Oncology, Veru, GlaxoSmithKline, Hoffmann-LaRoche AG/Genentech, Inc Travel, Accommodations, Expenses: Merck, AstraZeneca Dr. Nancy Davidson: No Relationships to Disclose
A real-world study reported at the ESMO Breast Cancer 2023 Annual Congress identified a risk for inappropriate therapeutic decision-making resulting from an alarmingly high rate of false-negative tests coming from biopsy specimens looking for biological parameters such as PR and HER2. Study researchers at the University of Catania suggested tumor heterogeneity was a big reason why markers like PR and HER2 were being missed that subsequently turned up in surgical specimens. But biopsy technique was also an issue, they concluded. OncTimesTalk correspondent Peter Goodwin caught up with one of the study authors, Federica Martorana, Assistant Professor of Medical Oncology. Together with lead author Sabrina Nucera from the University of Messina, and their co-authors, she said they retrospectively analyzed data from samples—collected over a 10 year period—for which both biopsy and surgical specimens were available. They excluded patients receiving neoadjuvant therapy and looked for differences in the detection of estrogen and progesterone receptors plus Ki67, as well as HER2 score, tumor grade, and intrinsic subtype. As a result, they had to re-classify 25 out of 61 cases of luminal B-like breast cancer that turned out mostly to be luminal A, with three of them being triple-positive.
Escuchar en Spotify: La Dra. Teresa Alonso, Oncóloga Médica del Hospital Universitario Ramón y Cajal en Madrid, España, nos comenta sobre los últimos resultados en supervivencia libre de progresión y supervivencia global del estudio fase III NETTER-1 evaluando Lutecio Lu-177 dotatate vs. octreotide LAR con escalamiento de dosis en pacientes con TNE de origen en el intestino medio y un Ki67 ≤ 20% que progresaron a análogos de la somatostatina. Adicionalmente, la Dra. Alonso nos presenta los resultados del estudio fase II OCCLURANDOM, el cual evalúa Lutecio Lu-177 dotatate vs. sunitinib en pacientes con TNE pancreáticos en primera línea. Fecha de grabación: 10 de marzo 2023
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2023.03.22.533784v1?rss=1 Authors: Chovatiya, G., Li, K. N., Ghuwalewala, S., Tumbar, T. Abstract: Adult skin homeostasis involves global reorganization of dermal lineages at different stages of the mouse hair growth cycle. Vascular endothelial cadherin (VE-cadherin encoded by Cdh5) expressing cells from blood and lymphatic vasculature structures are known to remodel during the adult hair cycle. Here we employ single-cell RNA-sequencing (scRNA-seq) 10x-genomics analysis of FACS-sorted VE-cadherin expressing cells marked via Cdh5-CreER genetic labeling at resting (telogen) and growth (anagen) stage of hair cycle. Our comparative analysis between the two stages uncovers a persistent Ki67+ proliferative EC population and documents changes in EC population distribution and gene expression. Global gene expression changes in all the analyzed populations revealed bioenergetic metabolic changes that may drive vascular remodeling during HF growth phase, alongside a few highly restricted cluster-specific gene expression differences. This study uncovers active cellular and molecular dynamics of adult skin endothelial lineages during hair cycle that may have broad implications in adult tissue regeneration and for understanding vascular disease. Copy rights belong to original authors. Visit the link for more info Podcast created by Paper Player, LLC
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2023.03.03.531045v1?rss=1 Authors: Adelman, J. W., Rosas, S., Schumacher, M., Mokry, R., Terhune, S. S., Ebert, A. Abstract: Human cytomegalovirus (HCMV) is a highly prevalent viral pathogen that typically presents asymptomatically in healthy individuals despite lifelong latency. However, in 10-15% of congenital cases, this beta-herpesvirus demonstrates direct effects on the central nervous system, including microcephaly, cognitive/learning delays, and hearing deficits. HCMV has been widely shown to infect neural progenitor cells, but the permissiveness of fully differentiated neurons to HCMV is controversial and chronically understudied, despite potential associations between HCMV infection with neurodegenerative conditions. Using a model system representative of the human forebrain, we demonstrate that induced pluripotent stem cell (iPSC)-derived, excitatory glutamatergic and inhibitory GABAergic neurons are fully permissive to HCMV, demonstrating complete viral replication, competent virion production, and spread within the culture. Interestingly, while cell proliferation was not induced in these post-mitotic neurons, HCMV did increase expression of proliferative markers Ki67 and PCNA suggesting alterations in cell cycle machinery. These finding are consistent with previous HCMV-mediated changes in various cell types and implicate the ability of the virus to alter proliferative pathways to promote virion production. HCMV also induces significant structural changes in forebrain neurons, such as the formation of syncytia and retraction of neurites. Finally, we demonstrate that HCMV disrupts calcium signaling and decreases neurotransmission, with action potential generation effectively silenced after 15 days post infection. Taken together, our data highlight the potential for forebrain neurons to be permissive to HCMV infection in the CNS, which could have significant implications on overall brain health and function. Copy rights belong to original authors. Visit the link for more info Podcast created by Paper Player, LLC
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2023.02.12.528218v1?rss=1 Authors: Li, A., Yi, J., Li, X., Dong, L., Ostrow, L. W., Ma, J., Zhou, J. Abstract: Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease that affects all voluntary muscles in the body, leading to paralysis and respiratory failure, while the extraocular muscles (EOMs) are largely spared even at the end-stage of ALS. Through whole-mount muscle imaging, we detected severe denervation along with depletion of Pax7+ satellite cells (SCs) peri-neuromuscular junction (NMJ) in hindlimb and diaphragm muscles of end-stage SOD1G93A mice (a familial ALS mouse model), but not in EOMs. Upon isolating SCs from different muscles using fluorescence activated cell sorting (FACS), the FACS profiles of hindlimb and diaphragm SCs of G93A mice exhibited activation and depletion phenotypes but not in wildtype controls. Importantly, both wildtype and G93A EOM SCs exhibited spontaneous activation behavior without significant differences in abundance. Examination of Pax7+ and Ki67+ cell ratios and RNA-Seq of SCs cultured in growth and differentiation medium revealed that EOM SCs maintained renewability and stemness better than diaphragm and hindlimb counterparts, especially in differentiation-favoring environments. Comparative functional annotation analyses indicate enrichment of axon guidance molecules, such as Cxcl12, in cultured EOM SCs. In neuromuscular coculture experiments, overexpressing Cxcl12 in G93A hindlimb SC-derived myotubes enhanced motor neuron axon extension and improved innervation, partially replicating the multi-innervation property of EOM SC-derived myotubes. The unique SC homeostasis regulation and the production of axon guidance molecules including Cxcl12 may explain the ALS resistant nature of EOMs. Intriguingly, feeding G93A mice with sodium butyrate extended the life span of G93A mice, alleviated NMJ denervation and SCs depletion. Butyrate treatment promoted renewability and stemness of cultured G93A hindlimb and diaphragm SCs, as well as Cxcl12 expression. Thus, butyrate-induced EOM SC-like transcriptomic patterns may contribute to its beneficiary effects observed in G93A mice. Copy rights belong to original authors. Visit the link for more info Podcast created by Paper Player, LLC
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2023.02.09.527879v1?rss=1 Authors: Jensen, G. S., Beaulieu, A. N., Curtis, C. D., Passarelli, J., Blaszkiewicz, M., Thomas, S., Morin, T., Willows, J. W., Greco, C. W., Brennan, C. J., Aniapam, C., Caron, L., Alves, M. J., Lynes, M. D., Carlone, D. L., Breault, D. T., Townsend, K. L. Abstract: Telomerase reverse transcriptase (TERT) is expressed by quiescent adult stem cells (ASC) in numerous adult murine and human tissues, but has never been explored in the adult brain. Here, we demonstrate that TERT+ cells in the adult mouse brain represent a novel population of multipotent ASCs that are localized to numerous classical neuro/gliogenic niches (including the ventricular-subventricular zone, hypothalamus, and olfactory bulb), as well as more recently described regions of adult brain plasticity such as the meninges and choroid plexus. Using a direct-reporter mouse line, we found that TERT+ cells expressed known neural stem cell markers such as Nestin and Sox2, but not markers of committed stem/progenitor cells, nor markers of mature neuronal or glial cells. TERT+ ASCs rarely expressed the proliferation marker Ki67, and in vitro TERT+ cells lost TERT expression when activated by growth factors, together indicating a quiescent phenotype similar to what has been observed in other tissues. When cultured, TERT+ cells behaved like neural stem cells by forming neurospheres, which could proliferate and become more metabolically active once stimulated by growth factors. TERT+ cells were observed in numerous brain niches, particularly near the ventricles and cerebrospinal fluid barriers, but notably, TERT+ cells were never observed in the hippocampus. Lineage tracing of TERT+ cells in adult transgenic mice (mTERTrtTA::oTET-Cre::RosamTmG) revealed large-scale expansion of TERT+ progeny and differentiation to diverse cell types in multiple brain regions. For example, lineage-traced cells expressed markers of mature neurons, oligodendrocytes, astrocytes, ependymal cells, and choroid epithelial cells, thus demonstrating the striking multipotency of this stem cell population in basal tissue turnover of the adult brain. Together, these data demonstrate that TERT+ cells represent a novel population of multipotent stem cells that contribute to basal plasticity and regeneration in the adult mouse brain. Copy rights belong to original authors. Visit the link for more info Podcast created by Paper Player, LLC
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2023.02.08.527694v1?rss=1 Authors: Prina, M., Esteve-Perez, R., Navarro-Moreno, C., Abellan-Alvaro, M., Lanuza, E., Sanchez-Catalan, M. J., Martinez-Garcia, F., Agustin-Pavon, C., Torres-Perez, J. V. Abstract: In mammalian species, motherhood is a critical period of neural plasticity, allowing the dam to adapt its behaviour to the demanding task of taking care of the pups. For example, in mice, pregnancy and lactation promote proliferation of neuroblasts in the subventricular zone and adult neurogenesis in the olfactory bulb. Thus, we hypothesised that the non-canonical neurogenesis described in other areas of the olfactory system would be similarly affected by either pregnancy or lactation. To test this hypothesis, we analysed the expression of doublecortin (DCX) and Ki67, markers of immature neurons and proliferation respectively, across the reproductive cycle in adult CD1 female mice. In a group of late pregnant females, there was a trend towards increase of DCX cells in the olfactory bulb. In the piriform cortex, lactating females had significantly fewer immature neurons, and of lower complexity and diameter, than pup-sensitised females. This suggests that both lactation, and/or exposure to pups, and the preceding pregnancy are require to promote maturation of this population of embryonically generated DCX-ir neurons. By contrast, the number of DCX-immunoreactive cells in the olfactory tubercle was not significantly affected by motherhood. Finally, we found Ki67 nuclei located in close apposition to clusters of DCX-ir cells in this region, with some of DCX-ir neurons co-expressing both markers. Taken together, our results expand our current knowledge on how motherhood remodels the brain and reveal the olfactory tubercle as an unexplored niche for adult neurogenesis. Copy rights belong to original authors. Visit the link for more info Podcast created by Paper Player, LLC
Featuring perspectives from Drs Aditya Bardia, Matthew Goetz, Virginia Kaklamani, Kevin Kalinsky and Hope Rugo, including the following topics: Current Role of Genomic Assays for Hormone Receptor (HR)-Positive Localized Breast Cancer Introduction (0:00) Case: A premenopausal woman in her early 40s with 9-mm, Grade III, ER/PR-positive, HER2-negative, node-negative infiltrating ductal carcinoma (IDC) – 21-gene Recurrence Score® 22 — Alan B Astrow, MD (3:39) Case: A premenopausal woman in her mid 30s with 3.6-cm, ER/PR-positive, HER2-low (IHC 1+), sentinel node-positive (4/4) multifocal IDC after bilateral mastectomies, adjuvant AC-T and ovarian function suppression (OFS)/aromatase inhibitor, Ki67 50% — Laila Agrawal, MD (9:40) Dr Goetz presentation (19:43) Optimizing the Management of Localized ER-Positive Breast Cancer Case: A woman in her early 40s with 5.5-cm, ER/PR-positive, HER2-negative, node-positive (20/21) IDC after bilateral mastectomies, bilateral salpingo-oophorectomy, adjuvant AC-T and initiation of letrozole/abemaciclib, Ki-67 3% — Susmitha Apuri, MD (32:22) Case: A woman in her mid 50s with de novo ER-positive, PR-negative, HER2-negative ulcerated breast cancer with pulmonary and extensive spinal metastases — Jennifer L Dallas, MD (40:45) Dr Kaklamani presentation (45:32) Selection and Sequencing of Therapy for Patients with ER-Positive Metastatic Breast Cancer (mBC) Case: A woman in her early 50s with ER/PR-positive, HER2-low mBC with a PI3KCA mutation who experiences a dramatic response to rechallenge with fulvestrant and a CDK4/6i (abemaciclib); now with progression and cytopenias — Kapisthalam (KS) Kumar, MD (59:07) Dr Kalinsky presentation (1:09:53) Recent Appreciation of HER2 Low as a Unique Subset of HR-Positive Breast Cancer Case: A premenopausal woman in her late 30s with ER/PR-positive, HER2-low (IHC 1+) IDC after adjuvant tamoxifen and OFS x 5 years, now with bone and liver metastases — Dr Agrawal (1:19:45) Dr Bardia presentation (1:24:29) Novel Strategies Under Investigation for Patients with HR-Positive mBC Case: A woman in her early 90s with ER/PR-positive, HER2-low (IHC 1+) mBC and progressive disease on multiple lines of endocrine and chemotherapy receives T-DXd — Dr Astrow (1:38:44) Case: A woman in her mid 40s with ER/PR-positive, HER2-low (IHC 2+) mBC who has received fulvestrant/abemaciclib, now receiving exemestane/everolimus – ESR1 and PIK3CA mutations — Dr Dallas (1:44:15) Dr Rugo presentation (1:49:58) CME information and select publications
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2022.12.22.521710v1?rss=1 Authors: Yagi, S., Lieblich, S., Galea, L. Abstract: Adult neurogenesis in the dentate gyrus plays an important role for pattern separation, the process of separating similar inputs and forming distinct neural representations. Estradiol modulates neurogenesis and hippocampus function, but to date no examination of the effect of estradiol on pattern separation have been conducted. Here, we examined estrogenic regulation of adult neurogenesis and functional connectivity in the hippocampus after the spatial pattern separation task in female rats. Ovariectomized Sprague-Dawley rats received daily injections of vehicle, 0.32 g (Low) or 5 g (High) of estradiol benzoate until the end of experiment. A single bromodeoxyuridine (BrdU) was injected one day after initiation of hormone or vehicle treatment and rats were tested in the delayed nonmatching to position spatial pattern separation task in the 8-arm radial maze for 14 days beginning two weeks after BrdU injection. Rats were perfused 90 minutes after the final trial and brain sections were immunohistochemically stained for BrdU/neuronal nuclei (NeuN) (new neurons), Ki67 (cell proliferation), and the immediate early gene, zif268 (activation). Results showed that only high estradiol reduced the density of BrdU/NeuN-ir cells and had significant inter-regional correlations of zif268-ir cell density in the hippocampus following pattern separation. Estradiol treatment did not influence pattern separation performance or strategy use. These results show that higher doses of estradiol can reduce neurogenesis but at the same time increases correlations of activity of neurons within the hippocampus during spatial pattern separation. Copy rights belong to original authors. Visit the link for more info Podcast created by Paper Player, LLC
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2022.12.21.521533v1?rss=1 Authors: Montgomery-Song, A., Ashraf, S., Santerre, P., Kandel, R. Abstract: Senescence, particularly in the nucleus pulposus (NP) cells, has been implicated in the pathogenesis of disc degeneration, however, the mechanism(s) of annulus fibrosus (AF) cell senescence is still not well understood. Both TNF and H2O2, have been implicated as contributors to the senescence pathways, and their levels are increased in degenerated discs when compared to healthy discs. This the objective of this study is to identify factor(s) that induces inner AF (iAF) cell senescence Under TNF exposure, at a concentration that can induce senescence in NP cells, bovine iAF cells did not undergo senescence, indicated by their ability to continue to proliferate as demonstrated by Ki67 staining and growth curves and lack of expression of the senescent markers, p16 and p21. Unlike iAF cells, NP cells treated with TNF accumulated more intracellular ROS and secreted more H2O2. Following TNF treatment, only iAF cells had increased expression of the superoxide scavengers SOD1 and SOD2 whereas NP cells had increased NOX4 gene expression, an enzyme that can generate H2O2. Treating iAF cells with low dose H2O2 (50 M) induced senescence, however unlike TNF, H2O2 did not induce degenerative-like changes as there was no difference in COL2, ACAN, MMP13, or IL6 gene expression or number of COL2 and ACAN immunopositive cells compared to untreated controls. The latter result suggests that iAF cells have distinct degenerative and senescent phenotypes. To evaluate paracrine signalling, iAF and TNF-treated NP cells were co-cultured. In contact co-culture the NP cells did induce iAF senescence. Thus, senescent NP cells may secrete soluble factors that induce degenerative and senescent changes within the iAF. This may contribute to a positive feedback loop of disc degeneration, and these processes could include H2O2 and cytokines (TNF). Further studies will investigate if human disc cells respond similarly. Copy rights belong to original authors. Visit the link for more info Podcast created by Paper Player, LLC
HPV testing plays an important role in cervical cancer screening, but not every patient whose HPV-positive will develop cervical cancer. One option that can help us identify which HPV-positive patients are truly at risk is CINtec PLUS Cytology, an immunohistochemistry test that simultaneously detects p16 and Ki67 biomarker proteins. Learn more about how this dual staining technique works in this video.
HPV testing plays an important role in cervical cancer screening, but not every patient whose HPV-positive will develop cervical cancer. One option that can help us identify which HPV-positive patients are truly at risk is CINtec PLUS Cytology, an immunohistochemistry test that simultaneously detects p16 and Ki67 biomarker proteins. Learn more about how this dual staining technique works in this video.
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2022.09.30.510371v1?rss=1 Authors: Yagi, S., Wen, Y., Galea, L. Abstract: Estrone and estradiol differentially modulate neuroplasticity and cognition but how they influence maturation pathways of new neurons is not known. The present study assessed the effects of estrone and estradiol on various aspects of neurogenesis in the dentate gyrus (DG) of ovariectomized young adult Sprague-Dawley rats using daily subcutaneous injections of 17{beta}-estradiol or estrone. Rats were injected with a DNA synthesis marker, 5-bromo-2-deoxyuridine (BrdU), and were perfused one, two, or three weeks after BrdU injection and treatment. Immunofluorescent labelling for Sox2 and Ki67 were used to examine the density of neural stem cells and proliferating cells, respectively. Double-immunofluorescent labelling of BrdU with doublecortin (DCX) or NeuN was used to examine the attrition and maturation of adult-born neurons over time. Estradiol reduced the density of neural stem cells in the dorsal DG, whereas estrone reduced the density of neural stem cells in the ventral DG. Furthermore, estradiol enhanced, whereas estrone reduced, cell proliferation after one week but not after longer exposure to hormones. Both estrogens increased the density of BrdU/DCX-ir cells after one week of exposure but showed greater attrition of new neurons between one and two weeks after exposure. Lastly, estradiol decreased the percentage of BrdU/NeuN-ir cells in the dorsal DG after three weeks of treatment. These results demonstrate that estrogens have differential effects to modulate several aspects of adult hippocampal neurogenesis in the short term, but fewer effects after long-term exposure and that estradiol and estrone modulate neurogenesis via different pathways. Copy rights belong to original authors. Visit the link for more info Podcast created by PaperPlayer
Introduction Mast cell tumors (MCTs) arise from malignantly transformed mast cells. In dogs, most of these tumors arise as primary tumors in the skin. They are the most common skin tumor in dogs, accounting for roughly 20% of all reported skin tumors.1 Any breed may be affected with MCTs, but certain breeds are predisposed, including golden retrievers, Labrador retrievers, Boston terriers, boxers, and pugs. Pugs are more likely to have multiple MCTs at diagnosis (56% of pugs with MCT in one study), but these tumors demonstrate more benign behavior and rarely lead to death.2 MCTs can affect dogs of any age but typically affect middle-aged to older dogs. An underlying etiology for most tumors cannot be identified. Breed predilections support some component of underlying genetic causes. Mutations in the c-kit tyrosine kinase receptor, which can lead to malignant transformation of mast cells, are found in 25%–30% of intermediate to high-grade tumors.3,4 KIT mutations will be further discussed in regards to both prognosis and treatment options for MCTs. MCTs can be located anywhere on the body and may lie within the dermis and/or subcutis. They have a wide range of gross appearance, from raised and superficial to very deep and fixed; they may feel soft and fluctuant or firm. Most MCTs are easily diagnosed with fine needle aspiration (FNA). Infrequently, MCT granules will not stain with Diff-Quik (Jorgensen Laboratories Inc., Loveland, CO, USA) and need to be stained with a Wright's stain. On Diff-Quik cytology, if eosinophils are seen along with large round cells that lack granules, suspicion should be raised for an MCT and the slide submitted to a clinical pathology laboratory for a non-Diff-Quik stain.5 The majority of MCTs will be cured with surgical excision.1 Prognostic factors for predicting MCTs that will exhibit a more aggressive biologic behavior – ie, tumors that will not be cured despite local excision and that will ultimately lead to the patient's death – are varied as well as controversial. When to pursue staging tests in dogs with MCTs, which tests to perform, and treatment recommendations beyond surgery are based on the predicted biologic behavior of the tumor, with staging diagnostics and systemic therapy the recommendation for dogs with biologically aggressive MCTs. Prognostic factors relating to history and physical examination Some factors that can be obtained from a history and physical examination that are generally accepted to carry a more guarded prognosis in dogs with MCTs include recent, rapid tumor growth and fixed, ulcerated tumors.6–8 Although publications regarding these features are limited, one early study reported doubling of the survival percentage at 30 weeks post-MCT excision for dogs with slow-growing tumors versus (vs) those with more rapidly growing masses.8 Biologically, both the ability to grow quickly and to become fixed to deeper tissues are physical manifestations of more aggressive behavior. Tumor location on the body can also be associated with biologic behavior; this topic is more controversial and the pertinent locations and published papers are highlighted as follows. Mucocutaneous location In limited published cases, eyelid margin MCTs appeared to have relatively benign behavior and were effectively treated with local therapy, although one dog was reported to have regional lymph node (LN) metastasis.9–11 MCT of the conjunctiva may be of concern only locally, without reported metastasis in three dogs.12,13 In a paper evaluating chemotherapy for high-risk MCT patients, eleven dogs with mucous membrane MCTs (vulva, prepuce, conjunctiva, oral cavity) had significantly shorter median survival times (MST) than 50 dogs with MCTs of haired skin.14 However, a recent paper of 32 dogs with 33 conjunctival MCTs treated with surgery alone showed prolonged survival times, with only two dogs having local recurrence despite incomplete margins in 25 cases, and no dogs dying of mast cell-related disease.15 Muzzle/perioral/oral location Early case reports described aggressive behavior and local metastatic disease at diagnosis in two dogs with MCT of the lip; survival times were 6 months or less.16,17 Of five dogs with MCT of the tongue, two presented with LN and/or systemic metastasis, and two of the remaining three had postoperative local recurrence leading to euthanasia.18 Larger, more recent studies confirmed that MCTs on the muzzle, perioral mucocutaneous junction, or oral mucosa have a more aggressive biologic behavior, with increased risk of locoregional LN metastasis.19–21 The rate of documented metastasis to local (mandibular) LNs was 55%–59%, compared with a 2 years) for low grade (85 dogs). A retrospective study with 47 dogs attempted to validate the two-tier scheme; although some of the data reported were inconsistent (including no description of the MI of the cases), the high-grade cases did have significantly worse progression-free and overall survivals than the low-grade ones.31 Further studies validating this proposed system are warranted; currently, the author's university pathology department provides the standard grade, an MI, and the two-tier grade for every sample. Clinically, heavy reliance is placed on the MI for behavior prediction. Other assessments on biopsies – highlighting mitotic index As grade is so variable and subjective, many other prognostic factors, including DNA aneuploidy, c-kit-staining pattern, presence of c-kit mutations, microvessel density, Ki67, proliferating cell nuclear antigen, and MI, have been evaluated in an attempt to better predict the behavior of canine MCTs and to pick out the “bad” grade II tumors. Two groups have evaluated the MI (total number of mitotic figures counted in ten high-power fields; fields with the highest mitoses counted) and found that it is predictive of survival time, even within the grade II tumor category. The groups did identify different values for the index, with the first paper showing an MST of >70 months for an index score ≤5 and survival 5.32 The second group wrote a letter to the editor in response to the first paper and confirmed that high scores have very short survival times, with index scores broken down into three groups: MI =0, MST not reached; MI =1–7, MST =18 months; and MI >7, MST =3 months.33 The cutoff of MI ≥7 was subsequently adopted for the two-tier system. Although MCT histologic “prognostic panels” are offered by some laboratories, no publications have shown how these panels may provide additional benefit over the MI and grade for prediction of tumor behavior. Staging information In general, staging tests for asymptomatic dogs with cutaneous MCTs are extremely low yield. The test most often positive is regional LN aspiration. Cytologic assessment of the locoregional LN is important, even if the node is not enlarged. In a study of dogs with muzzle MCT, four of eleven LNs with metastases were of normal size.19 Another study with 55 dogs with confirmed LN metastasis and 35 dogs without metastasis showed a sensitivity of 71% and specificity of 54% for palpation as a predictor of metastasis.34 Sixteen of 35 dogs (46%) with normal size LNs on physical examination had metastasis, whereas in another study eight out of 21 (38%) normal size LNs showed metastasis.35 FNA of the LN is ideally performed prior to excision of the primary MCT, as surgical treatment of the tumor can produce confusing LN results due to local postoperative inflammation. As mast cells can be a normal feature in LNs, some MCT cases will not have a definitive answer on LN cytology. Criteria for LN involvement have been proposed and used in several subsequent reports.34–37 Prognostically, the implication of a metastatic LN is also a controversial topic. LN involvement has been associated with a worse prognosis in a number of studies.20,36–39 However, several papers also report long-term survival in dogs with LN involvement where the primary tumor and the LN are treated to achieve local control using surgery with or without radiation therapy.14,34,35,40,41 Chemotherapy was used in many of the reported cases as well, although the protocols varied, and the added benefit of chemotherapy after local control was achieved cannot be proven via the retrospective noncontrolled studies that currently exist. To summarize the information in the literature, LN involvement may carry a worse prognosis, yet dogs can still have prolonged survival with adequate treatment of the primary tumor and the metastatic node, with chemotherapy potentially having a benefit as well. Assessment of any locoregional LNs, whether normal sized or enlarged, with cytology or histopathology is critical to determine the stage of the tumor and appropriate therapy for the patient. Other staging tests are rarely positive and may have false positive results as well. In general, staging tests other than LN aspiration are recommended in patients who have negative prognostic factors associated with their MCT. Thoracic radiographs are indicated to evaluate the sternal LN if the mass is on the ventral abdomen, or to rule out other non-MCT diseases. MCTs metastasize so rarely to the lungs that radiographs are not indicated to evaluate for pulmonary spread. Buffy coat preparation to look for circulating mast cells is a quick and easy test, but it is both insensitive and nonspecific. The bone marrow may still be infiltrated in spite of a normal buffy coat, and dogs with skin disease, parvo virus, and nonmast cell illnesses often have positive buffy coats despite not having an MCT.42–44 Bone marrow evaluation for mast cell infiltration is positive in
An interview with Dr. Fabrice André from Institute Gustave Roussy in Paris, France, and Dr. Komal Jhaveri from Memorial Sloan Kettering Cancer Center in New York, NY, authors on “Biomarkers for Adjuvant Endocrine and Chemotherapy in Early-Stage Breast Cancer: ASCO Guideline Update.” This updated guideline provides precise guidelines on previously endorsed genomic assays and recommendations on the use of new biomarkers to guide endocrine therapy and chemotherapy in patients with ER-positive HER2-negative tumors, and for those with HER2-positive or triple-negative breast cancer. Read the full guideline at www.asco.org/breast-cancer-guidelines. TRANSCRIPT Brittany Harvey: Hello and welcome to the ASCO guidelines podcast series brought to you by the ASCO Podcast Network, a collection of nine programs covering a range of educational and scientific content and offering enriching insight into the world of cancer care. You can find all the shows including this one asco.org/podcasts. My name is Brittany Harvey, and today I'm interviewing Dr. Fabrice André from Institut Gustave Roussy in Paris, France, and Dr. Komal Jhaveri from Memorial Sloan Kettering Cancer Center in New York, NY, authors on “Biomarkers for Adjuvant Endocrine and Chemotherapy in Early-Stage Breast Cancer: ASCO Guideline Update.” Thank you for being here Dr. André and Dr. Jhaveri. Dr. Fabrice André and Dr. Komal Jhaveri: Thank you. Brittany Harvey: First, I'd like to note that ASCO takes great care in the development of guidelines and ensuring that the ASCO conflict of interest policies follow each guideline. The full conflict of interest information for this guideline panel is available online with the publication of the guideline in the Journal of Clinical Oncology. Dr. André, do you have any relevant disclosures that are directly related to this guideline. Dr. Fabrice André: No. Brittany Harvey: Thank you. And Dr. Jhaveri, do you have any relevant disclosures that are related to this guideline topic? Dr. Komal Jhaveri: No. Brittany Harvey: Great, then let's talk about the content of this guideline update. So, Dr. Jhaveri, what prompted the update to the biomarkers for adjuvant endocrine therapy and chemotherapy in early-stage breast cancer, and what is the scope of this guideline update? Dr. Komal Jhaveri: So, since the biomarker guideline published in 2016, there were several new publications that came out between January 2016 and October 2021 that provided perspectives on the use of some of the genomic assays broadly, or specifically in women based on menopausal status, age or number of lymph nodes. Additionally, new biomarkers such as programmed cell death receptor ligand 1 or PD-L1, stromal TILs, circulating tumor DNA, or circulating tumor cells, or new applications, like extended endocrine therapy have also been developed. So, this updated guideline provides precise guidelines on previously endorsed genomic assays and also provides recommendations on the use of new biomarkers to guide endocrine therapy and chemotherapy in ER-positive HER2-negative, HER2-positive, and triple negative breast cancer. Brittany Harvey: Great. Thank you for setting the stage for this guideline. So, then Dr. André, I'd like to review the key recommendations of the guideline for our listeners. So, starting with, which biomarkers should be used to guide decisions on adjuvant endocrine and chemotherapy for patients with newly diagnosed ER-positive HER2-negative breast cancer? Dr. Fabrice André: Thank you. So, when we are focusing on a newly diagnosed patient with ER-positivity HER-2 negative in the context of the question about deciding on chemotherapy. First, the 21 gene recurrence score should be used in patients with node-negative and in patients with 1-3 node positive who are postmenopausal. In premenopausal, this test will be used only in patients with node-negative. The second test that is recommended is the MammaPrint 70-gene signature. And here the recommendation is to use this test if the patient is older than 50 and is node-negative or 1-3 node-positive. If the patient is 50 years or younger, the clinician should not use this test. Then also a recommendation is that the clinician could use the 12 gene risk score EndoPredict if the patient is postmenopausal and has breast cancer node negative or 1-3 positive-node. And finally, for the genomic test for the patient, the doctor could use PAM50. If the patient is postmenopausal and has breast cancer that is node-negative. Something new in this recommendation is the recommendation that the clinician could use Ki67. If the patient is postmenopausal and has stage I-II breast cancer with very specific recommendations on the cut off for positivity. Brittany Harvey: Great. Thank you for reviewing those recommendations, Dr. André, and highlighting the new Ki67. So, then, in your introduction, Dr. Jhaveri, you discuss the application of assays for extended endocrine therapy. So, which biomarkers should be used to guide decisions regarding extended endocrine therapy for patients with ER-positive HER2-negative breast cancer? Dr. Komal Jhaveri: Thank you. So, a quick word about extended endocrine therapy. I think extended adjuvant endocrine therapy, which is beyond five years of primary endocrine therapy, has certainly demonstrated improved outcomes, however, with modest absolute benefit and added toxicity and tolerability issues. Furthermore, while extended endocrine therapy is endorsed by clinical practice guidelines, clear guidance on individualized approaches to optimize patient selection for prolonged endocrine regimens, also remains limited. And this really underscores the need for prognostic and predictive information from genomic assays that can help guide this important clinical question we face. One such example that has now borne out has been the breast cancer index and really collected evidence from now five studies supports the prognostic and predictive utility of this assay, such that if a patient has node-negative or one to three positive nodes, and has been treated with five years of primary endocrine therapy, to guide decisions about extended endocrine therapy, the clinician may offer Breast Cancer Index with either tamoxifen or aromatase inhibitors or tamoxifen, followed by an aromatase inhibitor. Brittany Harvey: Great, thank you, Dr. Jhaveri. So, then, Dr. André, which biomarkers should be used to guide decisions on adjuvant therapy for early-stage HER2-positive breast cancer or triple-negative breast cancer? Dr. Fabrice André: Well, first, we have to be sure that we are good at defining HER2-positive breast cancer and triple-negative breast cancer. So, it's always good to remind colleagues to have quality control assessment in place for HER2, estrogen receptor, and progesterone receptor stainings. So far, when it's about predicting the benefit of adjuvant chemo or trastuzumab, there is no genomic test that is recommended in patients with HER2-positive or triple-negative breast cancer. Brittany Harvey: Great, thank you. So, then, Dr. Jhaveri, what recommendations did the expert panel make regarding emerging biomarkers? Dr. Komal Jhaveri: When we're talking about emerging biomarkers, we're referring to stromal TILs, we're referring to PD-L1, circulating tumor cells, and circulating tumor DNA. And while there is a lot of emerging data, currently, clinicians should not be utilizing these new biomarkers to guide decisions about adjuvant endocrine therapy or chemotherapy, as we do not have sufficient evidence to support their use. Brittany Harvey: Great and that's important for clinicians to know. So, then finally, Dr. André, in your view, how will these updated guideline recommendations impact both clinicians and patients? Dr. Fabrice André: So, we know Komal mentioned this at the beginning that now there is a very large amount of new publications, new data coming every day in the field of cancer. So, the only way for clinicians to deliver the best treatments for patients is to have the right guidelines with the right methodology, and ASCO is really putting in place a very rigorous methodology to deliver guidelines. And this is the best way to be sure that all patients will have access to the best decision by the doctor. So guidelines are also here to decrease the disparities in terms of the expert decision, and any patient in the world can have access to the best decision, thanks to guidelines delivered by ASCO and all the colleagues of the panel that worked very hard to deliver these guidelines. Brittany Harvey: Excellent! Well, thank you both so much, and the entire expert panel for your work to update these guidelines, and for taking the time to speak with me today, Dr. André and Dr. Jhaveri. Dr. Komal Jhaveri: Thank you. Dr. Fabrice André: Thank you, Brittany. Brittany Harvey: Thank you to all of our listeners for tuning in to the ASCO Guidelines Podcast Series. To read the full guideline go to www.asco.org/breast-cancer-guidelines. You can also find many of our guidelines in interactive resources in the free ASCO guidelines app available in iTunes or Google Play Store. If you've enjoyed what you've heard today, please rate and review the podcast and be sure to subscribe so you never miss an episode. The purpose of this podcast is to educate and to inform. This is not a substitute for professional medical care and is not intended for use in the diagnosis or treatment of individual conditions. Guests on this podcast express their own opinions, experience, and conclusions. Guest statements on the podcast do not express the opinions of ASCO the mention of any product or service organization activity or therapy should not be construed as an ASCO endorsement.
This special episode of Inside the Lab is a recording of the recent ASCP webinar "Ki67 as a Prognostic and Predictive Biomarker in HR-Positive/HER2-Negative Early Breast Cancer" featuring Drs. Sunil Badve MD, FRCPath and Manali Bhave, MD, both from Emory University School of Medicine, Atlanta, GA, USA.
Immunohistochemistry PearlsIn Part 3 of our Heme Path series, we break down the basics of immunohistochemistry (IHC)1. What is Immunohistochemistry?A. A molecular technique that utilizes fluorescently-labeled probes or chromogenic reporter stains to identify populations of cells. The goal being to help identify clonalityB. Procedure entails taking thin slices of tissue and then using the probes to look for the presence (or absence) of cells specific to that probeC. Helps to identify specific proteins, the pattern and distribution of which can help identify what it is that we are looking at2. Pros of IHC: A. Can be performed on fixed specimen (vs. flow cytometry, which requires live cells) B. Provides an idea of micro-architecture of the sample (diffuse distribution of cells vs. clusters) in context3. Cons of IHC: A. More labor intensive than flow, therefore can take longer to result B. Need to have an index of suspicion for what you are looking for. Your search is only as good as the probes you use! 4. Examples of utility: A. Diagnosis of acute leukemia: For instance, staining for CD34, which is expressed on blasts, can help you determine the “blast percentage.” That is, you count up number of cells that stain for CD34 probe then divide by total number of cells in the sample, which gives blast percentage. B. Diagnosis of plasma cell dyscrasias: These disorders result in “sticky” bone marrows, such that it is hard to get a good aspirate. This means on flow, you may not get an accurate percentage. Using IHC helps you better estimate the numbers. C. Diagnosis of lymphoma: Staining for specific markers (for example CD20, CD19 for B-cell lymphomas) can help in diagnosis. Can also use IHC to determine KI67, which is the proliferation index. D. Diagnosis of metastatic cancers: For instance, if you see a lung mass but also note axillary lymphadenopathy, so you sample the lymph node because less invasive. How you do you know if the cells in the lymph node are representative of metastatic lung cancer cells? Use IHC to stain them!E. PDL-1 status: This is believed to be important marker to predict response to immunotherapy. References https://www.abcam.com/content/immunohistochemistry-the-complete-guide - Great description of process and images to showcase what IHC staining looks like (not endorsement of product) Please visit our website (TheFellowOnCall.com) for more information Twitter: @TheFellowOnCallInstagram: @TheFellowOnCallListen in on: Apple Podcast, Spotify, and Google Podcast
This week, the FDA approved new indications for two drugs. The first approval was for abemaciclib in combination with endocrine therapy (tamoxifen or an aromatase inhibitor) for the adjuvant treatment of adult patients with hormone receptor–positive, HER2-negative, node-positive, early breast cancer who are at high risk of disease recurrence and who also have a Ki67 score of 20% or higher. The next day, the agency approved pembrolizumab in combination with chemotherapy, with or without bevacizumab, for patients with persistent, recurrent, or metastatic cervical cancer whose tumors express PD-L1.To listen to more podcasts from ASCO, visit asco.org/podcasts.
How many times did you get annoyed when using non-intuitive digital pathology software? Have you already given up on digital pathology and image analysis or are you still looking for something powerful but easy to use? Today's guest, Tuomas Ropponen, the chief technology officer at Aiforia, is talking about the creation of a pathology image analysis platform whose core principles are “easy to use” and “accessible” - Aiforia. Aiforia stands for artificial intelligence (AI) for image analysis (IA) and it combines cloud-based access with supervised deep learning for pathology image analysis. This is where pathologists and computer scientists collaborate closely to create tools that empower pathologists and give them access to the state-of-the-art image analysis methods. Aiforia began as a teaching and telepathology platform for sharing whole slide images. But as soon as deep learning passed the reality check and started outperforming classical computer vision methods, Aiforia's team knew that they had to incorporate this method into their platform. It would change the way pathology was done. The decision about using supervised deep learning as the method of choice was based on the desire to supervise the teaching of the AI in a similar way as we supervise the teaching of students. Only by supervising and curating the inputs can we be sure that we get quality output. Teaching AI in a supervised manner happens through annotations, and annotations are a natural way that pathologists communicate. Pathologists have always marked areas of interest on the glass and later digital slides. It has always been the way to show others what is important on the tissue. Adapting annotations for supervised deep learning as a way of showing AI what is important was a natural progression of how pathologists work. Another core value at Aiforia is the close collaboration between pathologists and computer scientists. Those two groups currently work very closely together but fostering this open relationship and honest communication required a few iterations and a deeper understanding of each other's ways of working. For computer scientists, it was surprising that the pathology scoring and grading system often could not be directly reproduced by image analysis algorithms. For pathologists it was surprising that the algorithm results did not match their visual estimates from glass slides. The two groups had to sit together and start dissecting the pathology problems into smaller components as well as translating them into quantifiable tasks. Suddenly it became clear to everyone that the Ki67 quantification in the tumor consists of first detecting the tumor epithelium and later identifying and counting the Ki67 positive and Ki67 negative cells within the epithelium. When pathologists' fatty liver scores of 70 or 80% were nowhere close to the absolute pixel area of fatty vacuoles in the liver tissue of max 20% it became evident that pathologists were subconsciously normalizing their scores and spreading them on a 0-100% scale. Coming together and analyzing the discrepancies as a team revealed that pathologists' scores or estimates are often an imprecise and inconsistent benchmark to measure against. Everyone went back to the drawing board (or drawing tablet) and provided a more objective ground truth – annotations. This close collaboration of pathologists and computer scientists as well as involvement of user experience designers helps drive innovation at Aiforia while maintaining accessibility and ease of use. To learn more about AI for image analysis visit Aiforia's website or even better, let the team show you what this whole thing is about - book a demo.
With more than 170K total downloads and over 700 citations in scientific literature, QuPath is arguably the most popular open-source software for quantitative pathology and bioimage analysis. Today's podcast guest, Pete Bankhead, the author of QuPath, is taking us behind the scenes of his software creation.Even though Pete is now a senior lecturer in digital pathology at the University of Edinburgh, his digital pathology career actually started by accident. With an undergraduate degree in theology and a master's in computer science, he started working on bioimage analysis during his PhD in biomedical sciences. He began using open-source software for image analysis, which was an excellent and very efficient way to work with static bioimages. So, during his post doc work he tried to apply open-source software to pathology whole slide images (WSI), unfortunately without success…The pathology WSI were just too big - it was not even possible to efficiently open them with any openly available software. So he started his own software development – first by creating his own plugins for already available open-source programs, such as ImageJ. It sort of worked but not really… There was no way to coordinate the development and bring all his plugins together, so he started developing his own pathology WSI viewer. That worked, and in the process, he realized that building software tools himself gave him a lot more freedom to solve problems in a way tailored to the specific challenges of digital pathology. He dove deeper into the project and created what we now know as QuPath – the open-source software for digital pathology image analysis. During its development, the software evolved from a Ki67 quantification tool to a machine learning-powered, versatile image analysis software.Listen to the full episode to learn Who and what influenced Pete, why is QuPath open-source,why Pete fought for QuPath to stay open-source, and what are the most important features of QuPath. This episode's resources:The real background story to QuPath on TwitterDigitizing a photo album with QuPathQuPath docs QuPath annotation tweetorial QuPath YouTube channel QuPath user forum (also for other software) QuPath immunofluorescence multiplex support StarDist cell detection algorithm (deep learning-based) QuPath 2020 workshop “From Samples to Knowledge”Cite this paper if you are using QuPath for researchMost popular free open-source software programs for image analysis of pathology slides HistoQC, an open-source way to control the quality of pathology images
In this episode, Javier Cortes, MD, PhD; Stephen R. D. Johnston, MA, FRCP, PhD; and Sara Tolaney, MD, MPH, answer questions from an audience of healthcare professionals on topics related to leveraging CDK4/6 inhibitors for HR-positive/HER2-negative early breast cancer, including:Clinical role of Ki-67 testingCore biopsies for identifying potential benefit with neoadjuvant endocrine therapyCombining CDK4/6 inhibition with tamoxifenPresenters:Javier Cortes, MD, PhDHead, Breast Cancer ProgramIOB Institute of OncologyMadrid and Barcelona, SpainStephen R. D. Johnston, MA, FRCP, PhDProfessor of Breast Cancer MedicineBreast UnitDepartment of MedicineRoyal Marsden HospitalLondon, United KingdomSara Tolaney, MD, MPHAssistant Professor of MedicineHarvard Medical SchoolAssociate DirectorSusan F Smith Center for Women's CancerDirector of Clinical Trials, Breast Oncology Director of Breast Immunotherapy Clinical ResearchSenior PhysicianBreast Oncology ProgramDana-Farber Cancer InstituteBoston, Massachusetts, USAContent based on an online CME program supported by an educational grant from Lilly.Link to full program, including downloadable slides and associated Podcast Pearls PDF:https://bit.ly/37tfvtj
The PENELOPE-B phase III trial did not show a benefit to the addition of one year of the CDK4/6 inhibitor palbociclib to adjuvant endocrine therapy in patients with hormone receptor-positive/HER2-negative breast cancer and residual disease after neoadjuvant chemotherapy. TRANSCRIPT This JCO podcast provides observations and commentary on the JCO article “Palbociclib for Residual High-Risk Invasive Hormone Receptor-positive, HER2 negative Early Breast Cancer – The PENELOPE-B Trial,” by Sybille Loibl and colleagues. My name is Erica Mayer, and I am a clinical investigator at the Dana-Farber Cancer Institute in Boston, MA who specializes in breast cancer. Since the initial approval of the CDK4/6 inhibitor palbociclib in 2015, this class of agents has become a standard part of management for most patients with hormone receptor positive HER2 negative advanced breast cancer. Use of one of the available CDK4/6 inhibitors - palbociclib, abemaciclib, or ribociclib - in combination with endocrine therapy, improves progression-free, and even overall survival, in the advanced disease setting, as shown in the respective trial series PALOMA, MONARCH, or MONALEESA. Although differentiated by their toxicity profiles, with palbociclib and ribociclib contributing more neutropenia and abemaciclib more diarrhea, across-the-board the benefit gained from use of these agents is remarkably preserved between trials, without apparent differential drug activity. Following the observation of benefit from use of CDK 4/6 inhibitors in the advanced setting, clinical trials evaluating the three available agents were subsequently launched to evaluate these agents in the adjuvant setting, after prior treatment with surgery, chemotherapy, and radiation if necessary. The first two trials to report in 2020, PALLAS and MONARCH-E, enrolled patients with early hormone receptor positive HER2- breast cancer, randomizing them to receive a CDK 4/6 inhibitor, palbociclib or abemaciclib respectively, for a planned 2-year duration, in addition to ongoing adjuvant endocrine therapy, versus ongoing adjuvant endocrine therapy alone. Notably, PALLAS enrolled patients with stage II and III breast cancer, whereas MONARCH-E targeted a high-risk group identified by tumor size, nodes, and Ki67 status. Initial reports from these trials were presented at an interim follow-up of about 20-24 months, a time when many were still receiving the combination therapy. The MONARCH-E analysis demonstrated a benefit from the addition of abemaciclib to endocrine therapy in prolonging invasive disease-free survival. This benefit was not observed with the addition of palbociclib in the PALLAS trial. The ongoing NATALEE trial, offering 3-years of reduced dose ribociclib in a similar setting, continues accrual. It is within this context that we place the PENELOPE-B trial. This trial enrolled the highest risk patients with early HR+/HER2- breast cancer: those with residual disease after preoperative chemotherapy and a high CPS-EG score, which is a calculation of anatomic stage and tumor biology meant to identify patients at highest risk of cancer recurrence. A total of 1250 patients were randomized to receive either one year of adjuvant palbociclib with ongoing adjuvant endocrine therapy, or placebo with endocrine therapy, and followed for a primary endpoint of invasive disease-free survival. As expected, the study enrolled a very high-risk population, with many patients having at least T2 disease and 4 involved lymph nodes at time of surgery. At a median follow-up of 43 months, no difference in invasive disease-free survival was observed, with a 4-year iDFS of 73% in the intervention arm, versus 72.4% in the placebo arm. No clinicopathologic subgroup appeared to derive benefit from the research intervention. The toxicity experience was similar to that seen in the metastatic setting, with notable and expected hematologic toxicity, but no significant difference in overall non-hematologic adverse events. Overall, drug exposure was satisfactory, with about 17% of patients unable to complete the planned 1-year duration of therapy. The PENELOPE-B study makes its mark in the adjuvant CDK4/6 landscape in several important ways. First, PENELOPE-B has the greatest duration of follow-up, allowing analysis of mature data and outcomes during and following adjuvant CDK4/6 inhibition. Although it is tempting to interpret a slight visual divergence of the Kaplan Meyer curves during the palbociclib exposure, where there is an absolute difference in iDFS of 4.3% at 2 years, there is no statistical evidence that the 2-year iDFS estimates are different between the two arms, and therefore no evidence of time-dependent efficacy. Importantly, in regard to experiences within the other CDK4/6 inhibitor trials, which have reported at an earlier time point, longer-term follow-up of those trials will be necessary to determine stability and maturity of the observed results. Additionally, PENELOPE-B is the only placebo-controlled trial presented to date. For that reason, the toxicity data presented represents the purest description of the experience of receiving a CDK4/6 inhibitor in the adjuvant setting in a population with prior chemotherapy exposure. Finally, when discriminating among the CDK4/6 inhibitors study populations, it is important to remember that PENELOPE-B enrolled a population of greatest established risk, suggesting any apparent differential outcome among the trials is unlikely related to differing baseline risk of accrued patients. Overall, PENELOPE-B represents a significant contribution to the breast cancer field, and substantially adds depth and dimension to the exploration of CDK4/6 inhibitors in the adjuvant setting. At the time of publication, it is unclear if an approved role will emerge for a CDK4/6 inhibitor in adjuvant therapy for our early breast cancer patients. Many questions remain regarding patient/tumor selection, agent differentiation, duration of therapy, and potential biomarkers. Importantly, the correlative analyses planned for all of the CDK4/6 inhibitor trials and molecular exploration of patient samples will substantially improve our understanding of the impact of these drugs in the early setting, and our overall ability to treat this subset of breast cancer. While waiting for these data, we should continue our focus on providing the best possible care for our patients with hormone receptor-positive, HER2-negative breast cancer who are being treated with endocrine therapy. This concludes this JCO podcast. Thank you for listening.
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2020.11.19.390401v1?rss=1 Authors: Geread, R. S., Sivanandarajah, A., Brouwer, E., Wood, G., Androutsos, D., Faragalla, H., Khademi, A. Abstract: In this work, a novel proliferation index (PI) calculator for Ki67 images called piNET is proposed. It is successfully tested on four datasets, from three scanners comprised of patches, tissue microarrays (TMAs) and wholeslide images (WSI), representing a diverse multicentre dataset for evaluating Ki67 quantification. Compared to state of the art methods, piNET consistently performs the best over all datasets with an average PI difference of 5.603%, PI accuracy rate of 86% and correlation coefficient R = 0.927. The success of the system can be attributed to a number of innovations. Firstly, this tool is built based on deep learning, which can adapt to wide variability of medical images and it was posed as a detection problem to mimic pathologists workflow which improves accuracy and efficiency. Secondly, the system is trained purely on tumour cells, which reduces false positives from non-tumour cells without needing the usual pre-requisite tumour segmentation step for Ki67 quantification. Thirdly, the concept of learning background regions through weak supervision is introduced, by providing the system with ideal and non-ideal (artifact) patches that further reduces false positives. Lastly, a novel hotspot analysis is proposed to allow automated methods to score patches from WSI that contain significant activity. Copy rights belong to original authors. Visit the link for more info
Link to bioRxiv paper: http://biorxiv.org/cgi/content/short/2020.09.26.315036v1?rss=1 Authors: Bjerke, D., Li, R., Price, J. M., Dobson, R., Rodrigues, M., Tey, C., Vires, L., Adams, R. L., Sherrill, J. D., Styczynski, P. B., Goncalves, K., Maltman, V., Przyborski, S., Oblong, J. Abstract: Human skin is exposed daily to environmental stressors, which cause acute damage and inflammation. Over time this leads to morphological and visual appearance changes associated with premature aging. Topical vitamin A derivatives such as retinol (ROL), retinyl palmitate (RPalm), and retinyl propionate (RP) have been used to reverse these changes and improve the appearance of skin. This study investigated a stoichiometric comparison of these retinoids using in vitro and ex vivo skin models. Skin biopsies were treated topically to compare skin penetration and metabolism. Treated keratinocytes were evaluated for transcriptomics profiling and hyaluronic acid (HA) synthesis and treated 3D epidermal skin equivalents were stained for epidermal thickness, Ki67, and filaggrin. A retinoic acid receptor-alpha (RARalpha) reporter cell line was used to compare retinoid activation levels. Results from ex vivo skin found that RP and ROL have higher penetration levels compared to RPalm. RP is metabolized primarily into ROL in the viable epidermis and dermis whereas ROL is esterified into RPalm and metabolized into the inactive retinoid 14-hydroxy-4,14-retro-retinol (14-HRR). RP treatment yielded higher RARalpha activation and HA synthesis levels than ROL whereas RPalm had a null effect. In keratinocytes, RP and ROL stimulated similar gene expression patterns and pathway theme profiles. In conclusion, RP and ROL show a similar response directionality whereas RPalm response was inconsistent. Additionally, RP has a consistently higher magnitude of response compared with ROL or RPalm. Copy rights belong to original authors. Visit the link for more info
Activation of immune cells is the all-important first step in mounting an immune response. Immune cell activation is a popular area of research because so much happens that is key to the downstream goal of fighting infection, cancer, and disease. There are many ways to measure immune cell activation, and they all have utility. Methods can be grouped into four main categories: Proliferation Assays, Cytokine Measurement, Surface Antigen Expression, and Cytotoxicity. In this webinar, we'll discuss specific assays in each of these categories, the joys and pitfalls of each assay, and recommendations on how to choose the best method. You will learn tips and strategies for successful assay development using the following methods: Proliferation: - 3H-Thymidine Uptake - Bromodeoxyuridine Uptake (BrdU) - ATP Luminescence - Fluorescent Dye Reduction (CFSE) Cytokine Measurement: - Multiplex vs. Single Cytokine - Choice of Cytokine (IFNg, TNFa, IL-6, IL-1?, etc.) - Kinetics of Cytokine Release Surface Antigen Expression: - CD69, CD25, PD-1, etc. - Combine with CFSE, Ki67 or BrdU - Kinetics are Important Cytotoxicity: - Two-Label Flow Cytometry - Calcein AM Dye Release - Luciferase Transduced Targets - Annexin V
Dr Hurvitz speaks with ecancertv at SABCS 2016 about the outcomes of the neoMONARCH trial of abemaciclib, a CDK 4/6 inhibitor, in combination with anastrozole for women with HR positive breast cancer. The study found that, alone or with anastrozole, abemaciclib induced significantly greater cell cycle arrest, defined by reduced Ki67 expression.
Dr Hurvitz presents data at San Antonio Breast Cancer Symposium 2016 from the phase II neoMONARCH trial of abemaciclib, a CDK4/6 inhibitor, as a single agent or combined with anastrozole for post-menopausal women with HR , HER- breast cancer. The study found that, alone or with anastrozole, abemaciclib induced significantly greater cell cycle arrest, defined by reduced Ki67 expression.
Prof Bundred talks to ecancertv at SABCS 2015 about early results from the UK EPHOS-B trial on the effect of peri-operative anti-HER2 therapy on cancer cell proliferation and cell survival. The UK EPHOS-B trial is an ongoing, multicentre, two-part randomised trial in patients with operable newly diagnosed HER2 primary breast cancer. The Independent Data Monitoring Committee for the trial has agreed partial release of the data from the first part of the study only in which women about to undergo surgery were randomised to receive peri-operative treatment with lapatinib or trastuzumab alone, or to no treatment. With approximately 11 days’ lapatinib treatment, decreased cell proliferation (defined as a ≥30% fall in Ki67, a marker of cell proliferation) was measured in 67% of women. An increase in cell death (≥30% rise in apoptosis) was measured in 30% of women. The key point is that you need to know the HER2 status of women before undergoing surgery, Dr Bunsen suggests. This is not always done but if it is and women are positive it shows that giving them lapatinib as early as possible could significantly improve their outcome.
Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 16/19
Beim kolorektalen Karzinom treten Biomarker zunehmend in den Fokus. Ziel muss daher die Etablierung von effektiven Markern sein, um den Patienten eine möglichst effektive, auf das Individuum zugeschnittene Diagnostik und Therapie anbieten zu können. In unserer Studie zum kolorektalen Karzinom sind nach Ausschlusskriterien n=108 Patienten mit Primärtumoren und n=59 Patienten mit Lebermetastasen vom KRK in die statistische Analyse eingegangen. Aus postoperativ entnommenem, N2-schockgefrorenem Gewebe wurde ein Biomarker-Profil aus molekulargenetischen und immun-histologischen Markern erstellt. Genetische Mutationen auf dem KRas- und BRaf-Gen wurden mittels Mikrodissektion/PCR/Pyrosequenzierung detektiert und mittels immunhistochemischer Färbungen die Moleküle EGF-R, Her2/neu, IGF1-R, c-Met, CD44v6, Ki67, CD45 und HLA-DR analysiert. Die statistische Analyse erfolgte univariat mittels Chi-Quadrat-Test, T-Test, Mann-Whitney-U-Test und Kruskal-Wallis-Test. Überlebensanalysen erfolgten mittels Kaplan-Meier-Schätzer und Cox-Regressionsanalyse. Hierbei ergaben sich in der univariaten Analyse zwischen den einzelnen Parametern der Primärtumoren folgende statistisch signifikante Korrelationen: Eine Mutation/der Wildtyp im KRas-Gen korrelierte mit dem L-Stadium, einer Mutation/der Wildtyp im BRaf-Gen und IGF1-R (p=0,007; 0,003; 0,034). Der um die Mutation G13D erweiterte KRas Wild-typ bzw. mutiert (ohne G13D) korrelierte hierunter mit dem L-Stadium, der Histologie, sowie ebenfalls mit BRaf (p=0,038; 0,039; 0,011). Eine Mutation im BRaf-Gen (Exon 15) war mit dem Geschlecht, der Lokalisation und dem Grading des Primärtumors signifikant verbunden (p=0,042; 0,003; 0,002). Der EGF-R korrelierte mit der Lokalisation, dem Grading, dem L-Stadium und CD44v6-Positivität (p=0,031, 0,020, 0,006, 0,021). Bei Her2/neu fand sich für das Geschlecht, die Lokalisation, CD45 und für HLA-DR eine Ver-knüpfung (p=0,005, 0,021, 0,032, 0,006). Zudem konnte ein Zusammenhang zwischen Her2/neu (Score) und dem Geschlecht (p=0,009) sowie der Tumorlokalisation (p=0,010) nachgewiesen werden. Für c-Met und IGF1-R (p=0,021) und für IGF1-R mit der Lokalisation des Primarius (p=0,027) bestand eine positive Korrelation. Des Weiteren korrelierten CD45 mit HLA-DR (p=0,046) und Ki67 mit dem Alter der Patienten (p=0,015). Hinsichtlich der Lebermetastasen konnte eine Verbindung von EGF-R mit der Histologie, mit IGF1-R und c-Met (p=0,046; 0,004; 0,007) nachgewiesen werden. Zudem konnte für c-Met und der Tumor-größe, dem löslichen präoperativen Tumormarker CA19-9 und CD44v6 (p=0,004; 0,003; 0,044) eine positive Korrelation nachgewiesen werden. HLA-DR und der BMI wiesen einen statistisch signifikanten Wert (p=0,022) auf und für Ki67 und CD44v6 bestand ebenfalls eine signifikante Verbindung (p=0,007). Ki67 und das Grading (p=0,017) sowie Her2/neu (Score) und die Histologie der Lebermetastasen (p=0,018) wiesen ebenfalls Signifikanz auf. Für das Gesamtüberleben im Kollektiv der Primärtumoren konnte im Kaplan-Meier-Schätzer ein Zusammenhang von N-Stadium (p
Dr Torsten Nielsen talks to ecancer at SABCS 2012 about how the immunohistochemical assessment of the cell proliferation marker Ki67 is of interest for potential use in clinical management. Dr Nielsen explains that a lack in consistency between labs detracts from Ki67's value as a marker. A working group was assembled to devise a strategy for Ki67 analysis and identify procedures to improve concordance.
Breast Cancer
This podcast reviews the rationale behind evaluation of metformin as a potential anti-cancer agent in breast cancer and describes the findings of a recent window of opportunity trial looking at the impact of metformin upon proliferative indices in women with early breast cancer.
Krüppel-like factor 8 (KLF8) has only recently been identified to be involved in tumor cell proliferation and invasion of several different tumor entities like renal cell carcinoma, hepatocellular carcinoma and breast cancer. In the present study, we show for the first time the expression of KLF8 in gliomas of different WHO grades and its functional impact on glioma cell proliferation. In order to get information about KLF8-mRNA regulation qPCR was performed and did not reveal any significant difference in samples (n = 10 each) of non-neoplastic brain (NNB), low-grade gliomas (LGG, WHO°II) and glioblastomas (GBM, WHO°IV). Immunohistochemistry of tissue samples (n = 7 LGG, 11 AA and 12 GBM) did not show any significant difference in the fraction of KLF8-immunopositive cells of all analyzed cells in LGG (87%), AA (80%) or GBM (89%). Tissue samples from cerebral breast cancer metastasis, meningiomas but also non-neoplastic brain demonstrated comparable relative cell counts as well. Moreover, there was no correlation between KLF8 expression and the expression pattern of the assumed proliferation marker Ki67, which showed high variability between different tumor grade (9% (LGG), 6% (AA) and 15% (GBM) of Ki67-immunopositive cells). Densitometric analysis of Western blotting revealed that the relative amount of KLF8-protein did also not differ between the highly aggressive and proliferative GBM (1.05) compared to LGG (0.93; p
Tierärztliche Fakultät - Digitale Hochschulschriften der LMU - Teil 04/07
The incretin hormones GIP (glucose-dependent insulinotropic polypeptide) and glucagon-like peptide-1 (GLP-1) mediate the so-called incretin effect, which describes the phenomenon that glucose given orally causes a higher insulin response compared to an isoglycemic intravenous glucose load. The insulinotropic action of GIP is reduced to almost absent in type 2 diabetes patients, while the action of GLP-1 is vastly preserved. GIPRdn transgenic pigs were generated by lentiviral genetransfer to establish a large animal model to investigate the effects of an impaired insulinotropic action of GIP on glucose homeostasis. At the age of 5 months GIPRdn transgenic pigs showed a disturbed oral glucose tolerance going along with reduced insulin secretion. Eleven-month-old GIPRdn transgenic pigs exhibited an impaired intravenous glucose tolerance and reduced insulin secretion as well as a significantly reduced total β-cell volume compared to controls. In this work different age classes (11 weeks, 5 months, 1-1.4 years) of GIPRdn transgenic were investigated to obtain detailed data about physiological and morphological characteristics. To proof specifity of the GIPRdn GIP/Exendin 4 stimulation tests were carried out in 11 week-old GIPRdn transgenic pigs. The insulinotropic action of intravenously injected porcine GIP was impaired, while this of Exendin-4, a GLP-1 mimetic, was enhanced in GIPRdn transgenic pigs compared to controls. Marked alterations in the expression profile of the GIPR and the GLP-1R were excluded as no apparent differences of immunohistochemically stained pancreas sections for GIPR and GLP-1R were detectable between GIPRdn transgenic pigs and controls at any age group. The effects of the impaired insulinotropic action of GIP on glucose metabolism were investigated by oral and intravenous glucose tolerance tests. Eleven-week-old GIPRdn transgenic pigs exhibited significantly reduced oral glucose tolerance with a delay in insulin secretion compared to controls. The area under the insulin curve (AUC insulin) during the first 45 minutes following glucose load was 31% smaller in transgenic pigs compared to controls. The total amount of insulin secretion was not different between the two groups indicating that GIPRdn expression initially only interferes with the incretin effect. This was supported by the fact that intravenous glucose tolerance and insulin secretion in transgenic pigs were not different from controls. Five-month-old GIPRdn transgenic pigs exhibited a tendency towards reduced intravenous glucose tolerance and reduced insulin secretion in response to an intravenous glucose challenge. To determine the reason for the alterations in glucose metabolism quantitative stereological analyses of the pancreas were performed. In 11-week-old pigs, transgenic and control groups showed similar β-cell mass. However, pancreatic -cell mass was reduced by almost 40% in 5-month-old and by 60% in adult (1 1.4 years) GIPRdn transgenic pigs compared to controls. Furthermore, the cellular composition of the islets was analyzed by quantitative stereological investigations. The relative volumes of α- and -cells in the islets were increased in 1-1.4-year-old GIPRdn transgenic pigs but the absolute volumes of these non-β-cell populations were not different from those of age-matched controls. To investigate the reason for the reduced pancreatic β-cell mass in GIPRdn transgenic pigs, β-cell proliferation and apoptosis rate was determined performing a double-immunohistochemistry for insulin and the proliferation marker Ki67 and cleaved caspase-3, respectively. Eleven-week-old GIPRdn transgenic pigs showed significantly less Ki67 positive cell nuclei compared to controls, whereas proliferation rates in 5 month-old and 1-1.4-year-old GIPRdn transgenic pigs reached no statistical significance. No differences were shown in the apoptosis rates of GIPRdn transgenic pigs compared to controls at any age, although a trend of higher numbers of cleaved caspase-3 positive β-cells was visible in 1-1.4-year-old GIPRdn transgenic pigs. In conclusion, GIPRdn transgenic pigs exhibit a comparable situation as in type 2 diabetes mellitus patients like impaired insulinotropic action of GIP, disturbed glucose tolerance and reduced β-cell mass. Moreover, the results of this work demonstrate an essential role of GIP for the physiological expansion of β-cell mass. In this context GIPRdn transgenic pigs represent a valuable model for further investigations on type 2 diabetes mellitus including diet studies and therapeutic trials.
Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 09/19
Das Mammakarzinom ist eine Tumorerkrankung mit einem sehr heterogenen Krankheitsverlauf. Um den individuellen Krankheitsverlauf einer Patientin besser vorhersagen zu können und um individuelle Therapieentscheidungen treffen zu können, bedarf es prognostischer und prädiktiver Faktoren. Neben den etablierten Prognosefaktoren Tumorgröße, Lymphknotenstatus, Metastasierung und Grading werden momentan verschiedene biologische Faktoren untersucht. Darüber hinaus wird in letzter Zeit insbesondere die prognostische Bedeutung von disseminierten Tumorzellen im Knochenmark diskutiert. Das Ziel der vorliegenden Arbeit war, die prognostische Relevanz von den biologischen Faktoren HER2, Topoisomerase-IIalpha, Ki67 und p53 sowie deren Korrelation mit dem Nachweis von disseminierten Tumorzellen im Knochenmark bei Patientinnen mit Mamma-CA zu evaluieren. Insgesamt wurden Gewebeproben von 256 primären Mammakarzinomen mit bekanntem Knochenmarkstatus untersucht. Zunächst wurden aus den archivierten Gewebeblöcken sogenannte Tissue-Micro-Arrays (TMAs) hergestellt. An diesen TMAs wurde die Expression von HER2, Topoisomerase-IIalpha, Ki67 und p53 mit Hilfe immunhistochemischer Färbungen analysiert. Zusätzlich wurde das Tumorgewebe auf eine potentielle Genamplifikation von HER2 und Topoisomerase-IIalpha mittels Fluoreszenz-in-situ-Hybridisierung (FISH) hin untersucht. Es stellte sich heraus, dass die meisten dieser biologischen Faktoren untereinander korrelieren. Darüber hinaus konnte eine signifikante Korrelation zwischen der Überexpression von HER2 und einem kürzerem krankheitsfreien Überleben der Patientinnen nachgewiesen werden. Weitere Korrelationen zwischen den einzelnen Faktoren und dem Krankheitsverlauf wurden nur in einigen Subgruppen gefunden. Keiner der untersuchten Faktoren stellte sich jedoch als unabhängiger prognostischer Faktor bezüglich eines kürzeren krankheitsfreien bzw. metastasenfreien Verlaufs oder eines kürzeren Gesamtüberlebens heraus. Diese Ergebnisse deuten daher lediglich auf einen kausalen Zusammenhang zwischen den von mir untersuchten Tumorsuppressormolekülen, Proliferationsmarkern und Wachstumsfaktorrezeptoren hin. Die Expression von HER2, Topoisomerase-IIalpha, Ki67 und p53 auf dem Primärtumor korrelierte nicht mit dem Vorhandensein von Tumorzellen im Knochenmark. Die Dissemination von Tumorzellen ins Knochenmark scheint daher ein von den untersuchten Faktoren unabhängiger Prozess zu sein. Allerdings korrelierte der Nachweis von disseminierten Tumorzellen im Knochenmark mit einem kürzeren Gesamtüberleben der Patientinnen. Bisher gehört die Knochenmarkspunktion zwar nicht zur Routinediagnostik beim primären Mammakarzinom. Allerdings wird diese Untersuchung in einigen Zentren empfohlen. Die weitere Charakterisierung von disseminierten Tumorzellen im Knochenmark sowie die Evaluation von zirkulierenden Tumorzellen im peripheren Blut sind Gegenstand der aktuellen Forschung.
Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 08/19
Thu, 3 Apr 2008 12:00:00 +0100 https://edoc.ub.uni-muenchen.de/8515/ https://edoc.ub.uni-muenchen.de/8515/1/Zerzer_Maryam.pdf Zerzer, Maryam
Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 07/19
Besides of dental erosions, sialadenosis manifesting as bilateral swelling of parotid glands, is a typical manifestation of bulimia. Objective: The mechanism of acinar enlargement in sialadenosis is obscure, although peripheral polyneuropathy secondary to systemic disorders is assumed to be significant. This study aimed to find new aspects of pathogenesis of sialadenosis. Methods: Based on a fortuitous observation of diminished alpha-actin-positive myofilaments in myoepithelial cells in sialadenosis, 11 cases each of control and sialadenosis parotid glands were morphometrically analysed assisted by immunohistochemistry for alpha-actin, p63, cytokeratin 14 and Ki67 including double staining. Results: In sialadenosis: the acini were significantly (p
Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 07/19
Bei dem Kaposi-Sarkom (KS) handelt es sich um eine eng mit dem Humanen-Herpes-Virus 8 (HHV 8) assoziierte vaskuläre Neoplasie. Die Etablierung eines kommerziell erhältlichen, stabilen Antikörpers gegen das virale „Latenz-assoziierte-nukleäre Antigen“ (LANA) von HHV 8 ermöglicht den Nachweis HHV-8-infizierter Zellen an Ex-vivo- Paraffinmaterial. In dieser Arbeit wird diese Methode an einem Kollektiv aus insgesamt 61 Patienten mit HIV-assoziiertem KS angewendet, um einen Beitrag zur Beantwortung von bislang ungeklärten Fragen nach der quantitativen Verteilung von HHV 8 im Organismus und zur formalen Pathogenese des KS zu leisten. Das Kollektiv besteht aus diagnostischen Exzisaten (20 Früh- und 20 Spätstadien des KS) sowie aus 21 Sektionsfällen mit KS wovon Lympknoten, Leber-, Milz- und Lungengewebe untersucht wurden. Durch die Etablierung und Auswertung technisch aufwendiger immunhistochemischer Doppelfärbeexperimente mit dem viralen Protein LANA und verschiedenen strukturellen und regulatorischen Humanproteinen (CD31, CD34, Ki67 und p21) sowie durch die Anpassung des LANA-Nachweises an Autolyse- bzw. Sektionsmaterial kommen wir in der vorliegenden Arbeit zu folgenden Ergebnissen: Die KS-Progression geht von der Proliferation nicht-infizierter Zellen aus. Dies spricht für ein parakrin-stimuliertes KS-Wachstum und stützt Hypothesen, die das KS als viral-getriggerte, reaktive Angioproliferation einstufen. Außerdem unterscheidet dieser Befund das KS von dem ebenfalls HHV-8-assoziiertem „Primären Effusions Lymphom“ (PEL), dessen Progression ein autonomer Zellzyklus zugrunde liegt. Weitere Ergebnisse dieser Arbeit stützen die mittels In-vitro-Experimenten von Wang et al. aufgestellte Hypothese, dass HHV 8 das Expressionsmuster der infizierten Zellen in Richtung der endothelialen Vorläuferzelle verändert. Durch Koexpressionsexperimente mit Endothelzellmarkern kann außerdem gezeigt werden, dass die HHV-8-infizierten Zellen an der Gefäßbildung im KS direkt beteiligt sind. Schließlich lassen sich im Sektionsmaterial HHV-8-infizierte mononukleäre Zellen in mehreren Organen als disseminierte Einzelzellen außerhalb der klinisch/makroskopisch sichtbaren KS-Läsionen nachweisen, und es finden sich mikroskopisch kleine KS-Frühstadien. Der bisher nur in vitro durch den Nachweis von HHV-8-infizierten Zellen im Blut vermutete formalpathogenetische Weg, wonach HHV-8-infizierte mononukleäre Zellen zur Infektion der endothelialen KS- „Zielzellen“ beitragen, wird somit ex vivo an dem Untersuchungskollektiv histomorphologisch belegt.
Tierärztliche Fakultät - Digitale Hochschulschriften der LMU - Teil 03/07
Insulin-like growth factors (IGF) are not only mediators of metabolic actions, but also regulate cell growth, differentiation and apoptosis. IGF-II is of particular interest because of its mitogenic effects. It is known that endothelial progenitor cells (EPC) improve myocardial function after acute myocardial infarction. The aim of this study was to investigate whether overexpression of IGF-II in EPC further contributes to improvement in left ventricular function after myocardial infarction. Human CD34+ cells from cord blood were isolated and cultured in adequate cell medium. Early passage EPC were transduced ex vivo by a retroviral vector expression of IGF-II (EPC-IGF-II) or empty vector (EPC-pLXSN) and expanded up to 46 population doublings. Expression levels were confirmed by RT-PCR. Athymic, nude rats were thoracotomized and ligation of the LAD (left anterior descending artery) was performed for 30 minutes before reperfusion was initiated. Vector only transduced EPC or EPC-IGF-II cells were injected immediately after reperfusion in the border of the infarct zone. Serial echocardiographic measurements were performed to analyze left ventricular ejection fraction (EF) up to 2 weeks after myocardial infarction when animals were mercy killed. Transplantation of EPC-derived cells improved left ventricular function after experimental myocardial infarction from 47,3 ± 1,8 % (EF of the control group, n = 11) to 51,4 ± 0,7 % EF 2 weeks after infarction (p < 0,05, n = 9). Overexpression of IGF-II further improved left ventricular ejection fraction to 53,6 ± 0,5 % EF at 2 weeks as compared to empty vector transduced cells (p < 0,05, n = 10). In vitro experiments revealed that IGF-II dose-dependently enhanced the proliferation capacity of H9C2 rat cardiomyoblasts measured by a BrdU incorporation assay. Immunhistological analysis of proliferating cells in the myocardium showed an increased number of Ki67+ cells within the infarct zone 7 days after transplantation of IGF-II overexpressing cells as compared to empty vector transduced cells. It was shown that transplantation of IGF-II overexpressing EPC impaired the infarction size by nearly 20 % in comparison to EPC-pLXSN (p < 0,05). Thus, transplantation of IGF-II overexpressing EPC in acute myocardial infarction may improve myocardial function by enhancing the proliferation capacity of resident cardiomyocyteprogenitors.
Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 05/19
Deregulation of epidermal growth factor receptor (EGFR) pathways contributes to the progression of a wide range of cancers, and the EGFR is an attractive therapeutic target. The humanized EGFR-specific monoclonal antibody EMD72000 has shown potent inhibitory activity in preclinical experiments and is under investigation in clinical studies. Using the human L3.6pl pancreatic cancer cell line, we investigated whether the efficacy of EMD72000 can be enhanced in combination with gemcitabine. Nude mice were orthotopically injected with L3.6pl cells followed by biweekly treatment of 40 mg/kg EMD72000 and 100 mg/kg gemcitabine (either alone or in combination). Under two treatment schedules we evaluated a) the influence of different time points of initiation of single agent and combined treatment after tumor cell injection into nude mice and b) the influence of different treatment durations. In the first experiment, treatment was initiated at four different time-points after tumor cell injection (days 8 - 28). Exposure to EMD72000 or gemcitabine alone resulted in detectable tumor shrinkage and reduced lymph node and liver metastases, however, these effects were enhanced in the EMD72000 plus gemcitabine groups. Furthermore, combination treatment as well as EMD72000 monotherapy led to a significant reduction of microvessel density and tumor cell proliferation in primary pancreatic tumors following immunohistochemical analysis for CD31 and Ki67, respectively. Overall, the effects were strongest when treatment was initiated at early time points after tumor cell injection. In a second experimental set-up, treatment was initiated at day 8 after tumor cell injection in all cases and stopped at four different time-points (days 15 - 33). Interestingly, there was no significant difference in the average tumor weight of the groups treated for short versus longer time periods. The treatment was most effective when given shortly after tumor cell injection, whereas the duration of the treatment appeared less important in this model. In conclusion, in this model the anti-angiogenic and anti-tumor activity of EMD72000 in combination with gemcitabine was substantially more effective than either treatment alone.
Pathogenic events leading to in-stent restenosis (ISR) are still incompletely understood. Among others, inflammation, immune reactions, deregulated cell death and growth have been suggested. Therefore, atherectomy probes from 21 patients with symptomatic ISR were analyzed by immunohistochemistry for pathogen burden and compared to primary target lesions from 20 stable angina patients. While cytomegalovirus, herpes simplex virus, Epstein-Barr virus and Helicobacter pylori were not found in ISR, acute and/or persistent chlamydial infection were present in 6/21 of these lesions (29%). Expression of human heat shock protein 60 was found in 8/21 of probes (38%). Indicated by distinct signals of CD68, CD40 and CRP, inflammation was present in 5/21 (24%), 3/21 (14%) and 2/21 (10%) of ISR cases. Cell density of ISR was significantly higher than that of primary lesions ( 977 +/- 315 vs. 431 +/- 148 cells/mm(2); p < 0.001). There was no replicating cell as shown by Ki67 or PCNA. TUNEL+ cells indicating apoptosis were seen in 6/21 of ISR specimens (29%). Quantitative analysis revealed lower expression levels for each intimal determinant in ISR compared to primary atheroma (all p < 0.05). In summary, human ISR at the time of clinical presentation is characterized by low frequency of pathogen burden and inflammation, but pronounced hypercellularity, low apoptosis and absence of proliferation. Copyright (C) 2004 S. Karger AG, Basel.