Tag Archive for: clinical trials

Expert Insights on Overcoming Barriers to Myelofibrosis Care

Expert Insights on Overcoming Barriers to Myelofibrosis Care from Patient Empowerment Network on Vimeo.

How is myelofibrosis care impacted by barriers, and what are solutions for healthcare providers to overcome them? Experts Dr. Raajit Rampal from Memorial Sloan Kettering Cancer Center and Dr. Jeanne Palmer from Mayo Clinic discuss different access barriers that impact quality of care and ways that healthcare providers can help close disparity gaps for patients.

See More from EPEP MPNs

Related Resources:

Expert Insights on Overcoming Barriers to Myelofibrosis Care

Overcoming Barriers in Myelofibrosis Care: Challenges Faced by Patients and Providers

HCP Roundtable: Breaking Through Myelofibrosis Practice Barriers

HCP Roundtable: Breaking Through Myelofibrosis Practice Barriers

Explaining Myeloproliferative Neoplasm Disease Progression to Patients

Explaining Myeloproliferative Neoplasm Disease Progression to Patients


Transcript: 

Dr. Nicole Rochester:

So we’re going to start by diving into the nuanced challenges and practice barriers in myelofibrosis care. We know that some of those factors may be related to socioeconomic factors and health disparities. So, Dr. Rampal, I’m going to start with you. What are the primary barriers in myelofibrosis care, and how might these barriers impact accessing effective treatment for myelofibrosis?

Dr. Raajit K. Rampal:

Well, I think it’s broad in the sense that, if we think about access to care, I think that part of the problem lies in underdiagnosis, and that is really on the end of the spectrum of access to primary care I’d say, are people getting in and getting regular blood count checks, the things that are going to tip somebody off that, you know, if somebody has a hematologic issue or problem, I think that is a big part of the entire discussion and spectrum here. And then the second thing is that this is a rare disease, and there are a handful of specialists throughout the world who deal with this.

And so making sure that patients have access to expert care, which is not to undermine the quality care that’s provided in the community, but this is more a question about do patients have access to, you know, clinical trials or to the most updated knowledge, and that to me really revolves around people being able to have access to tertiary care referral center who has a myelofibrosis expert. So I think those are some of the barriers, at least in my mind.

Dr. Nicole Rochester:

Thank you so much. And, Dr. Palmer, I’d love to get your insight as well. From your perspective, what are the primary barriers to accessing effective treatment, and what strategies can be implemented to overcome these barriers?

Dr. Jeanne M. Palmer:

So I agree with everything that Dr. Rampal said, but I’d also want to add to it, like many people would have to travel a distance to see that specialist. And so one of the ways that I’ve been able to try to overcome that thus far is by telemedicine. So being able to have that ability to contact somebody over the Internet, I think, especially if they’re getting very good care locally, just being able to provide that sort of expert additional advice about how to manage their disease, what different options are available.

Fortunately, this is a space where new drugs are coming pretty rapidly. So I think that having the ability to be able to weed through all of these different drugs, understand the pros and cons of them, and advise patients is good. And if they can’t make it to see you, then they can’t get advice about the medications nor can the providers. Because recognizing this is such a rare disease, a community provider has a lot to keep track of. So trying to keep track of something that impacts so few patients is hard to do. I think the other big barrier is cost and support for getting these medications.

These medications are all, unfortunately, quite expensive, and new drugs that are coming down the horizon will probably be so as well. So being able to find the right support for them, even when insurance covers it, it’s sometimes with a very large copay. So trying to handle access to these drugs, not only in the knowledge of which drugs to give, but also the ability to be able to pay for them.

Dr. Nicole Rochester:

Wonderful. Thank you both for elucidating some of those barriers. And you both mentioned that this is a rare disease and, Dr. Rampal, you also talked about expert care. And so I’d love to know, and I’ll start with you, Dr. Rampal, how do referral patterns impact treatment access in myelofibrosis and particularly for underserved populations?

Dr. Raajit K. Rampal:

This is a really interesting question, because things are changing in the sense that, I think at least in the New York area, but probably true elsewhere, there’s increasing consolidation of healthcare as hospital systems buy up smaller practices. And that means that referral patterns are going to change and are changing. There’s more of an impetus for docs to refer patients within their own health network. And they may or may not have access to expert care within their network. I think that’s one thing to keep in mind.

And the second is that, the elephant in the room here is that you have to have insurance to get into these networks. You have to have the right insurance. And do all of these big academic tertiary centers accept every kind of insurance? The answer is no, they don’t. So right off the bat, you have a systemic barrier, but then with the changing referral patterns, and I think likely certain insurances being more likely to be accepted in certain networks, you’ve already kind of fragmented the entire system. So, is there a streamlined way for patients to get in? Right now, I think the answer is no, there are a lot of barriers.

Dr. Nicole Rochester:

Thank you, Dr. Rampal. Dr. Palmer, do you have anything to add? And also if you can touch on what healthcare providers should be aware of and what they can pay close attention to with regard to these barriers that you and Dr. Rampal have discussed?

Dr. Jeanne M. Palmer:

Right, so I completely agree with everything he said. I think it’s a real challenge, especially as you parse apart these different healthcare systems. I think one of the important things for patients to know and what providers can help with is providing sort of access to some of the patient advocacy sites. On these patient advocacy sites, they can find the name of different providers, and sometimes that helps them call in to get a referral. Now, the insurance coverage is another challenge that’s a lot harder to manage. But I think one thing that patients can do is if within their own network, there isn’t an expert, at least being able to go to these patient advocacy sites, finding out who they should, who they can go see.

Dr. Nicole Rochester:

Wonderful, thank you. Well, we’ve been talking about how certain populations may experience more barriers. And certainly we know that’s at the root of health and healthcare disparities. So I want to move and start to talk a little bit about cultural competency. And I’ll go back to you, Dr. Rampal, what role do you believe cultural competency plays in addressing barriers in myelofibrosis care?

Dr. Raajit K. Rampal:

Yeah, I think one of the major tasks is to be aware that cultural competency is important. That to me is sort of the first step in everything. And in making this a little bit more granular. Not every patient approaches their disease or their diagnosis in the same way. And a lot of that is informed by their cultural beliefs, their community. And this is something, you know, living in a place in like New York where we see people from all different cultures, this is a striking thing because there are people who, for example, you know, they come from, you know, strong religious faith backgrounds. Their approach to things is different.

In some respects, they approach the disease and the need for treatment in a very different way than people who come from other cultural communities or those who are, let’s say, even not coming from a religious setting. But if you’re not aware of those and you try to put the same sort of treatment paradigm on all patients, you’re going to run into conflicts at some point. So I think to me, the first step is to be aware that these things influence how people perceive their disease, how they perceive the treatments or their desire to even be treated. But if you’re not aware of those things at the outset, then you’re going to run into those issues, I think.

 


Share Your Feedback

Advice for Setting Myeloma Treatment Goals and Collaborating on Care Decisions

Advice for Setting Myeloma Treatment Goals and Collaborating on Care Decisions from Patient Empowerment Network on Vimeo.

This animated video defines common myeloma treatment goals, provides an overview of available myeloma therapies, and includes advice for making care decisions and the importance of being your own advocate.

See More from Collaborate Myeloma

Related Resources:

Collaborate | Being an Empowered Myeloma Patient

Understanding Myeloma | How You Can Collaborate in Your Care

Understanding Myeloma | How You Can Collaborate in Your Care

The Benefits of Shared Decision-Making for Myeloma Care


Transcript:

Bianca: 

Hi! I’m Bianca, a nurse specializing in myeloma. In this video, we’ll discuss myeloma therapy and explain how to work with your healthcare team so that you can choose a care plan best suited for YOUR myeloma. 

I’d also like to introduce you to Suzanne. Suzanne is a patient advocate living with myeloma.  

Suzanne: 

Thanks, Bianca. I’m happy to share my own experience and to talk about how I worked with my healthcare team to decide on a care plan. 

When my doctor and I were first considering my options, we started by setting treatment goals. Bianca, can you define treatment goals?  

Bianca: 

Sure! Each patient is unique, so it may vary by person. You should collaborate WITH your healthcare team to determine YOUR treatment goals. Common goals of myeloma treatment may include: 

  • Reducing and managing your symptoms. 
  • Slowing the progression of the disease. 
  • Inducing remission. 
  • And, helping you live longer while maintaining quality of life.

Suzanne: 

Thanks for explaining this. When I discussed treatment goals with my doctor, I pointed out that I still wanted to be able to play pickleball with my friends, and to care for my young grandchildren. We reviewed options that could allow me to stay as active as possible and manage my myeloma at the same time. 

Bianca: 

That’s a great point, Suzanne. It’s also important to remember that your treatment goals can change throughout the course of your myeloma. Discuss them regularly, not just with your care team but also your care partner – that can be a family member or even a close friend. 

Suzanne: 

That’s right. I discussed my care with my husband AND my adult children – it’s important to keep your support team in the loop.  

And it’s also essential to understand the treatment options available to you. Bianca, what are the types of therapy available to treat myeloma? 

Bianca: 

There are a number of different classes of therapy, which include: 

  • Proteasome inhibitors; 
  • Immunomodulatory therapies or IMiDs; 
  • Monoclonal antibodies; 
  • Stem cell transplants; 
  • There are also the new and recently approved therapies such as bispecific antibodies and CAR (Chimeric Antigen Receptor) T-cell therapy; 
  • And, of course, clinical trials.   

Clinical trials can be a good option for patients at any stage of disease, often giving patients early access to a viable and cutting-edge therapy. When considering treatment, you should ask your doctor if there is a clinical trial that may be right for you.  

Suzanne: 

Good point! When I was deciding on a therapy, my doctor and I, along with my husband, discussed the risks and benefits of each approach as well as the potential outcome of each option. My doctor also walked through the clinical trials that were available to me. 

Bianca, given all of the options, how do you decide which therapy is appropriate for a particular patient? 

Bianca: 

That’s a great question. As mentioned in our previous video, results of in-depth testing, which determine if a patient has low-risk or high-risk myeloma, can affect your choices. Other factors that impact treatment decisions may include: 

  • Your age, overall health, and any pre-existing conditions. 
  • Potential side effects of the treatment. 
  • Previous therapies that may have been used to treat your myeloma. 
  • The financial impact of a treatment plan. 
  • And the patient’s lifestyle and preference.  

Suzanne: 

That’s right. And, you shouldn’t hesitate to weigh in on what YOUR preference is. Do your own research so you understand each treatment approach, then work WITH your healthcare team to determine what might be best for you.  

Bianca: 

That’s excellent advice, Suzanne. So, when making treatment decisions, you should: 

  • Work with your healthcare team to understand your treatment goals. 
  • Talk with your doctor about all available treatment options. 
  • And, inquire about any clinical trials that may be right for you. 
  • Then, discuss the pros and cons of each option. 
  • Bring a care partner with you to appointments and take time post-visit to discuss the proposed plan and treatment options. 
  • And, always remember that you have a voice in your care. Speak up and ask questions. You are your own best advocate. 

Suzanne: 

Great advice, Bianca. Don’t forget to visit powerfulpatients.org/myeloma to learn more. Thank you for joining us!  

HCP Roundtable: Breaking Barriers and Cultivating Clinical Excellence in Endometrial Cancer Care

HCP Roundtable: Breaking Barriers and Cultivating Clinical Excellence in Endometrial Cancer Care from Patient Empowerment Network on Vimeo.

What are obstacles faced by healthcare providers (HCPs) treating endometrial cancer patients and families? How can HCPs foster a culture of innovation and continuous improvement in clinical practice? Dr. Radhika Gogoi of Karmanos Cancer Institute and Dr. Charlotte Gamble of MedStar Health unravel the complexities of endometrial cancer care, providing HCPs with the insights and tools needed to navigate challenges effectively and to deliver optimal care to their patients and families.

See More from EPEP Endometrial Cancer

Related Resources:

Dr. Charlotte Gamble: Why Is It Important for You to Empower Patients

Dr. Charlotte Gamble: Why Is It Important for You to Empower Patients?

Addressing Disparities in Gynecologic Oncology | Key Challenges and Solutions

Addressing Disparities in Gynecologic Oncology | Key Challenges and Solutions

Overcoming Barriers: Empowering Underrepresented Groups With Endometrial Cancer

Overcoming Barriers: Empowering Underrepresented Groups With Endometrial Cancer

Transcript:

Dr. Nicole Rochester:

Welcome to this Empowering Providers to Empower Patients or EPEP Program. I’m Dr. Nicole Rochester, founder and CEO of Your GPS Doc. EPEP is a Patient Empowerment Network program that serves as a secure space for healthcare providers to learn techniques for improving physician patient communication and to overcome practice barriers. In this endometrial cancer healthcare provider roundtable, we are discussing breaking barriers and cultivating clinical excellence in endometrial cancer care. 

This program aims to improve healthcare provider awareness of screening and access disparities to specialized care in endometrial cancer, while also addressing solutions to overcome practice barriers such as lack of awareness, outdated practices, and inertia. Today, we’ll talk about some of the complexities of endometrial cancer care and we’ll provide healthcare providers with the insights and tools needed to navigate challenges effectively.

Our discussion will cover enhancing healthcare provider awareness of diagnostic and access disparities to specialized care in endometrial cancer, actionable strategies to overcome practice barriers in endometrial cancer care and empowering providers through fostering a culture of innovation and continuous improvement in clinical practice. 

It is my privilege to be joined by Dr. Radhika Gogoi from Karmanos Cancer Institute. Dr. Gogoi is a dedicated clinician and cancer researcher focused on exploring the Hippo pathway in gynecologic cancers, aiming to uncover novel therapeutic approaches. Thank you so much for joining us today, Dr. Gogoi.

Dr. Radhika Gogoi:

Thank you, Dr. Rochester, and thank you for having me. It’s great to be here.

Dr. Nicole Rochester:

It’s also my honor to be joined by Dr. Charlotte Gamble, gynecologic oncologist at MedStar Washington Hospital Center and MedStar Southern Maryland Hospital. Dr. Gamble is dedicated to taking care of patients who have historically been marginalized and are vulnerable to poor health outcomes. Her research examines the role of safety net hospital systems in gynecologic cancer care. Thank you so much for joining us today, Dr. Gamble.

Dr. Charlotte Gamble:

Thank you so much for having me. It’s a pleasure to be here.

Dr. Nicole Rochester:

So we’re going to start today’s discussion by diving into enhancing healthcare provider awareness of diagnostic and access disparities in endometrial cancer care. And I want to start by just framing the current situation. Black women are twice as likely to die from endometrial cancer when compared to their white women counterparts.

There is no current screening test for endometrial cancer, and diagnosis is usually made after patients present with symptoms. Sadly, the list doesn’t end there. So I’m going to start with you, Dr. Gogoi. What are the primary barriers to accessing specialized care for endometrial cancer that you’ve observed in your practice and perhaps in others?

Dr. Radhika Gogoi:

So thank you for that question. I guess I just want to start by just level setting a little bit and talking about specifically endometrial cancer disparities. So unlike other gynecologic cancers, which actually have been shown to be decreasing in incidence, endometrial cancer is actually one of the cancers that is increasing. We know that low grade endometrial cancers really have an excellent prognosis, but higher grade endometrial cancers really have a much poorer prognosis.

And that’s the specific subgroup that seems to be increasing in all women. Black women, again, as you mentioned, have the lowest survival rate, and that is even when corrected for the specific type of endometrial cancer and the stage of endometrial cancer. So with that sort of background and problem, the question really becomes how do we allow and educate our patients about the barriers that they face when accessing specialized care?

And so some of the barriers, at least that I’ve noticed, and certainly in the hospital that I practice in is really as you pointed out, that there is no good current screening test. Black women tend to have a delayed onset from the time of their symptoms, which in this case is really postmenopausal bleeding to actually obtaining a diagnosis.

And there are studies that have shown that some of that is education or there is an understanding that perhaps postmenopausal bleeding is not as significant an issue, doesn’t lead necessarily, to obtaining healthcare which as you can appreciate then delays the onset of the diagnosis. There is also that Black women present with more advanced disease. This is, again, likely due to the delay in diagnosis and the delay from diagnosis to getting treated.

Dr. Nicole Rochester:

Thank you. Thank you, Dr. Gogoi for level setting and for getting us started with a better understanding of those disparities. And I’d love to go to you, Dr. Gamble. In your experience, what are the primary barriers to addressing specialized care for endometrial cancer?

Dr. Charlotte Gamble:

Yeah, thank you so much.  I’ll just add some additional context. I think there are so many places in which these barriers can happen. So as Dr. Gogoi alluded to, sometimes that can happen at the patient level due to just not recognizing that having irregular, unpredictable bleeding, bleeding after menopause, bleeding even before menopause has happened, but really heavy bleeding or heavy periods that any sort of abnormal, heavy irregular bleeding is not normal and has to be evaluated in a timely fashion. But that’s at the patient level.

And sometimes, people have competing priorities where they might recognize that it’s a problem but not be able to make it to their doctor’s office. Have other kinds of things that are happening in their day-to-day lives where they just are not able to prioritize their own health. We also see then how the system can affect that.

If someone hasn’t had a gynecologist in years, or their gynecologist said, bye, you don’t need to see me anymore because you’re over the age of 65, you no longer need pap smears and they’ve fallen out of care, or patients who don’t have health insurance or patients who live really far away from their doctors. Accessing the healthcare system in the year of our Lord 2024 is actually really, really hard. If you lack the resources or lack the wherewithal to navigate that.

Additionally, what we see on the healthcare standpoint is that maybe patients do actually access the healthcare system. They call the gynecologist’s office. They call their primary care doctor, and they’re told by an admin staff or someone else that hears them, but that says, okay, fine, we’ll get you in, but it’s going to be in about three-and-a-half months.

And so sometimes those barriers and those delays come from the health system in general, which is also a challenge. And then even after they present to their doctor, sometimes they’re told, “Okay, let’s go ahead and let’s get an ultrasound first, and based on what your ultrasound looks like, then we’ll decide if we need to do a uterine biopsy to diagnose you.” But we also know that for certain types of these uterine cancers, specifically the really aggressive ones, that sometimes their ultrasound might look totally fine, but there still can be cancer underlying there.

And so I think that there are multiple barriers to getting even. That’s before the diagnosis even happens much less what comes after the diagnosis is had, how one gets from their gynecologist to a surgical subspecialist called the gynecological oncologist that Dr. Gogoi and I, this is our field. And there are multiple barriers and referral pathways there. But that’s to give a little bit more context that these things might start at the patient level, but the healthcare system, unfortunately, can contribute in rarely challenging ways to the barriers that patients face.

Dr. Nicole Rochester:

Thank you both for that. I think we have a really good idea of the breadth of this issue, and I appreciate both of you pointing out that there are patient level factors, but there are also system level factors that come into play. And, Dr. Gamble, you just kind of ended by talking about referrals.

So I’m going to pick up there. And let’s talk about referral patterns. And I’ll start with you for this question. How do those referral patterns impact access to specialized care gynecologic oncologists like yourself and Dr. Gogoi for women who are diagnosed with endometrial cancer and specifically for those underserved populations?

Dr. Charlotte Gamble:

Yeah, this is a complicated question and multi-layered. Again, I think big picture referral patterns. We know that there are barriers for patients who live rurally. We know there are barriers for patients who don’t have insurance or their insurance might be, they might be underinsured. And so there’s kind of system level barriers that we see on the macro level in some of our referral pathways.

And occasionally, sometimes patients with gynecologic cancers will actually be referred to not gynecologic oncologists, which are surgical subspecialists that work specifically with gynecologic malignancies, but might be referred to a general surgeon first, or a general gynecologist who may or may not know kind of what they’re getting into in terms of the care. I think what I’ve noticed in real life is that when I’m getting referrals, sometimes I’m getting a text message, sometimes I’m getting an email, sometimes getting a message in the medical record system, and the referrals are coming in many different ways.

Occasionally, patients are calling themselves. And so the ways that referrals actually then happen in real life and how those spread to kind of the macro levels is really interesting to me. It can be very complicated, very complex, and I think this is where the role of having healthcare navigators comes into play where folks can really assist patients in getting to the right surgical subspecialists.

Dr. Nicole Rochester:

Absolutely. And we know there’s a lot of evidence for the role of healthcare navigators in other disease spaces and specifically in cancer. So I appreciate you mentioning that. We’ve talked a little bit about patient level factors, and we’ve talked about system level factors. We haven’t talked a lot about maybe some provider level factors.

And I want to talk about cultural competency and the role that that plays in addressing disparities in endometrial cancer, specifically with regard to diagnosis. And I’m going to start with you, Dr. Gogoi on this one. And if you can share the role that you believe cultural competency plays in addressing some of these disparities that we’ve been talking about.

Dr. Radhika Gogoi:

Yeah, so I think you heard from us a little bit that oftentimes some of the symptoms that our patients have had are either ignored, or patients haven’t had a chance to fully address them, or the providers haven’t fully evaluated kind of the importance of their symptoms. And I think, so again, I think nurse navigators community educators are essential in sort of educating our communities about the importance of seeking care when you have sort of the symptoms that Dr. Gamble talked about and in a really timely fashion. I think that that’s really important as well.

So I think it obviously, cultural competency is important in sort of all facets of a patient’s journey as they kind of have the diagnosis through their treatment. But I think that the other place is really once they enter the healthcare system being aware and mindful of sort of healthcare mistrust both from the provider, from the institution, from a much larger governmental level and being aware of that distrust and really trying to take time to address their concerns.

That specifically at least comes up a lot in terms of clinical trials where we know that minority populations, Black patients are met vastly underrepresented. And so I think specifically in that area, there is really the opportunity to do a much better job in providing transparency about the trial design, about recruiting specifically minority patients. Interestingly, a lot of these landmark trials actually didn’t include race as a measure. And so we know that they’re underrepresented, but we actually don’t know how much minority populations are underrepresented. So important to really understand where patients are coming from when they enter a healthcare institution.

Dr. Nicole Rochester: 

Absolutely. Thank you. Thank you so much for that. Dr. Gamble, can you speak to awareness? You spoke about the fact that sometimes patients are referred to perhaps the wrong provider. You know, there was that sent, you mentioned to a general surgeon instead of to a gynecologic oncologist. And so we can imagine that there may be some challenges both in the primary care setting and perhaps even among general obstetrician gynecologists. So can you speak to how we can elevate the level of awareness to enhance healthcare provider awareness of diagnosis and access disparities and really appropriate referral patterns?

Dr. Charlotte Gamble:

Yeah, absolutely. I think it’s a tough question. I think it’s, again, like all things, it’s a little bit nuanced. Sometimes, again different levels of providers, different parts of the country, everybody practices a little bit differently. So everyone knows once you get a cancer diagnosis, you should probably go to a cancer specialist. And so generally, that’s coming to a gynecologic oncologist generally, or that’s sometimes that’s going to a medical oncologist. But occasionally, sometimes patients will have symptoms where it’s just abnormal bleeding, and they’re seeing a general gynecologist who then does a hysterectomy, and there’s a surprise diagnosis of an endometrial cancer.

Usually they’re, hopefully, they’re not making it too much to kind of the general surgery pathway our general surgery colleagues are awesome, but it’s kind of a different kettle of fish and the type of hysterectomy that’s needed and the type of specific surgery that’s needed to include lymph node assessment is different for somebody who’s getting a cancer surgery for uterine cancer compared to, let’s say, for fibroids or for adenomyosis or a non-gynecologic cancer situation.

I think, again, I’m on the receiving end of all of this, so I see patients who have made it to my doorstep and gotten kind of through the hoops and the barriers, but there’s definitely those out there that we know from the data somehow ended up with their surgery, not exactly in the appropriate hands.

And it’s hard to say, I don’t know if I actually even have advice for like how this is supposed to happen. I think we need to understand kind of the as Dr. Gogoi alluded to earlier, just kind of how prevalent endometrial cancer is right now and how the rates are rising and that abnormal bleeding has to be taken very seriously. And the thing that I harp on the most is a normal ultrasound does not mean there’s nothing else to explore there. There has to be a tissue biopsy.

And really impressing that on both patients as well as the first kind of people that they see, either their primary care doctor or even a nurse practitioner, a physician assistant. Just because there’s a normal ultrasound does not mean that your work is done. And we have studies to really demonstrate how that can delay patient’s care and missed diagnoses can happen. So it’s hard to answer your question, to like, how do we fix the referral pathway system? I don’t know the answer to that, and maybe Dr. Gogoi can speak to that, but I will say from like a screening standpoint, since there’s no great screening test, an ultrasound is not, might be the first step, but it’s definitely not the only step and it cannot be the last.

Dr. Nicole Rochester:

I appreciate that. As you were talking, I was thinking like this needs to be a major awareness campaign, not just for healthcare providers, but for patients as well. You know, this idea that abnormal or or postmenopausal bleeding is never normal and make sure your doctor gets a tissue biopsy and not just an ultrasound. Dr. Gogoi, we’ve talked already about some of the obstacles and barriers faced by patients. Can you talk about some of the obstacles or barriers that are faced by healthcare providers when treating endometrial cancer?

Dr. Radhika Gogoi:

Yeah, I think that the one other barrier that sort of comes up a lot specifically in terms of maybe even in terms of getting a patient to their referring providers or a G1 oncologist is transportation. I find that transportation is a huge issue for our patients. And it’s mostly associated with cost. So I think that one of the things that really, that we can do as a community is to somehow provide means or ways for our patients to get to us once they’re diagnosed. It’s hard enough to get them to us but when they’re facing challenges of cost and transportation, that becomes even more challenging.

So I think that that’s from a patient standpoint. And then, and then I think what Dr. Gamble mentioned about really the barriers for providers is really education around ultrasound and how those how specifically perhaps even in Black women, that it’s not as predictive of endometrial cancer risk and that an ultrasound alone is not enough of a workup for a number of these patients to rule out some underlying pathology. So that’s huge, and again mistrust of the medical profession is something that all healthcare providers deal with. And allowing time for patients to ask their questions, to be transparent about what it is that you’re doing and why I think goes a long way towards overcoming those challenges.

Dr. Nicole Rochester:

Thank you both for that. I think we’ve gotten a lot of information around the causes of the disparities and some actions that we can begin taking. Are there any unforeseen or outdated practice-related barriers that may hinder your work and that of your colleagues? And on that same note, are there any solutions or actions related to those? And I’ll start with you on this one, Dr. Gamble.

Dr. Charlotte Gamble:

Well, starting all the way back, I think an outdated practice is not listening to patients, and not recognizing your own privilege as a provider. I think that to Dr. Gogoi’s point that she’s mentioned a couple of times now, I have innumerable patients that just felt that they were not listened to felt that they couldn’t…that their stories were not being heard, that they faced no’s, no’s, no’s at multiple levels in trying to seek care for abnormal bleeding, and come to me very frustrated and kind of weary. And I think just really understanding how vulnerable patients are when they’re seeking care, and being cognizant of the privilege that we have as healthcare providers is something that is more contemporary compared to an outdated practice of being very paternalistic.

I guess sometimes I have had patients who are like, yeah, I was told that I needed a biopsy and I just…the way that they told me and the way that they said that it had to be done tomorrow, it freaked me out, and I couldn’t do it. And it’s just kind of unfortunate when the recommendations might be correct, but the way in which it’s being delivered is not being received by the patients in a way that they’re able to be receptive to.

I think another outdated practice again that I harp on that I’m such a strong believer in is like just the role of the ultrasound. I really, I don’t like it. I hate it. I think it’s useful for knowing uterine size and maybe if there’s some kind of stuff inside the uterus, but abnormal bleeding either before menopause or after menopause has to have a tissued biopsy. So I think that’s outdated to just get an ultrasound and have the patient come back, because it continues to perpetuate delays in care, and I think disparities to an extent. Another outdated…no, I don’t know. Is there anything else, Dr. Gogoi, that you’d add to this?

Dr. Radhika Gogoi:

I feel like this is a lot lot older now. It used to be at one point that we used to do all our endometrial cancer patients with open surgery. I think that now, I guess it’s even more outdated than outdated. Most of the surgeries are now pretty much done either minimally, invasively laparoscopically or robotically. There are obviously extenuating circumstances to both of those things, but I think approach to surgery is also sort of evolved.

The treatment paradigm for patients with advanced endometrial cancers has evolved even more so in the last maybe two years or so with the role of immunotherapy. So encouraging our patients to be educated about their options, both for route of surgery, for treatment and to make sure to ask those questions at the time of their office visit I think is something that we should all encourage.

Dr. Charlotte Gamble:

I would dovetail from that and also say, to kind of harken back to some of the other things we discussed in terms of referral pathways and things like that, and just how gynecologic oncology care is delivered in different parts of the country, there are various models for this, and so classically, patients who have gynecologic cancers, including endometrial cancer are managed solely by the gynecologic oncologist, meaning a surgical subspecialist like myself or Dr. Gogoi, who not only do the surgery, but also deliver if patients need it chemotherapy or specialized systemic therapies afterwards, sometimes with a component in partnership with a radiation oncologist.

But there are parts of the country by institution or by geography where the care that comes after the initial surgery done by the gynecologic oncologist might be shared with a medical oncologist who may or may not actually have super sub-specialized training when it comes to gynecologic cancers, because again, historically, this has been an area that’s been managed really by our subspecialty.

And so I encourage patients as well as referring doctors, et cetera, et cetera, to make sure that whoever, if there’s additional therapies that are needed after the hysterectomy is done, such as chemotherapy or immunotherapy, as Dr. Gogoi alluded to, that that is done in partnership with a gynecologic oncologist, because our field is kind of driving where so much of the contemporary understanding of how to manage these conditions long-term is coming from. And to make sure that if a medical oncologist is doing this, they feel very competent. They do this all the time, and they’re working in close partnership with their gynecologic oncology colleagues.

Dr. Nicole Rochester:

Wonderful. Thank you for that. So as we talk about outdated practices, I guess the other side of the coin is innovation and continuous improvement. And so I’ll turn this question to you, Dr. Gogoi. How can healthcare providers and hospital leaders foster a culture of innovation and continuous improvement? So that’s some of these outdated practices that you all, that you and Dr. Gamble, discussed are really no longer a thing?

Dr. Radhika Gogoi:

Yeah. So I couldn’t agree more.I think that some of those innovative approaches really come from enrolling in clinical trials. And so I think that the importance of having minority populations, equally represented in clinical trials is essential. Otherwise we’re not going to make the progress that we need to make to really equal the playing field here, so to speak. So I think that that really is a huge player trying to educate patients about clinical trial options, being transparent about what those look like, who benefits, what the data suggests, why it’s important to me is again, really key.

Dr. Nicole Rochester:

Wonderful. And Dr. Gamble, if we were to be specific, are there any healthcare provider to healthcare provider strategies, anything that you’ve seen work, maybe things that you’ve done yourself, innovative approaches or protocols with regard to this idea of innovation and continuous improvement?

Dr. Charlotte Gamble:

Yeah, I think different institutions have different cultures, and I think having institutional culture that embraces change that is, desires to be on the vanguard of advancing science, and the science could be in terms of bench research. It could be in terms of clinical trials, it could be in terms of healthcare delivery, really trying to advance and push forward. The science and how we get the best care to our patients in the most timely fashion I think is really important. Culture is hard to change, and we all work in institutions that have various different cultures. I think that healthcare provider to healthcare provider, I think open lines of communication are great. I tell folks, text, call, email, whatever you need to do to get patients in a timely fashion is necessary.

I think the understanding of we are one person working within a team is really necessary. So patients might say, oh, Dr. Gamble this, Dr. Gamble that, but I always, always, always tell them that I cannot do this work without my nurse navigator, without my fellow, without my residents, without my inpatient floor nurses, the oncology nurses, and really understanding that oncology care specifically is a really big team sport.

And healthcare leaders, when we’re thinking about things from a system level, I think sometimes have different like just help making sure that everybody understands how much of a team sport that this is. How much the radiation oncologists, the medical oncologists interface with us on a regular basis, I think is really important. And to understand that we’re all in this together to deliver the best care to our patients. I really think the role of health, like nurse navigators and lay navigators needs to be further pushed forward within our field, including not only gynecologic oncology, but just gynecology in general, and that’s some of the work that I’m doing from a research standpoint now. And so I’d love to see the role of navigators be fully embraced and somehow reimbursed by healthcare systems in general.

Dr. Nicole Rochester:

Absolutely. Well, it is time to wrap up this roundtable. I have really enjoyed talking with both of you as always. I have learned a lot. I’m sure that our audience will learn a lot from this conversation. And before we go, I’d love to get closing thoughts. So I’m going to start with you, Dr. Gogoi. What is one takeaway message that you would like to leave with the healthcare professionals who will watch this program?

Dr. Radhika Gogoi:

So I guess the one takeaway would be that, as I think about my role in sort of disparities prevention, if you say it is really as an educator, and so the importance of community education on symptoms, on diagnosis, on treatment approaches, I think it’s essential that we don’t think of ourselves as a silo. I’m not just, but I’m part of the larger community as Dr. Gamble spoke about. And to be part of that larger community means that I need to be within that community and function within the community. So whether it’s me, community navigators, health educators to really play a role in educating our patients about how to approach their symptoms.

Dr. Nicole Rochester: 

Wonderful. Thank you, Dr. Gogoi. And what about you, Dr. Gamble, what’s your closing thought that you would like to leave with the audience?

Dr. Charlotte Gamble: 

Yeah, I think for healthcare providers in general, I think the thing that I think is most important is, again, being able to listen to patients, working and encouraging the systems that we are housed into, be able to be responsive to them, so that if patients are educated, do know that there’s something wrong. They’re able to not only get a hold of someone, but also be listened to and heard and taken seriously. And we have a lot to do in that regard. So I think just understanding again, our role and how privileged we are to be in our jobs, in this line of work, and being able to leverage that to listen to patients and get them the timely care that they need.

Dr. Nicole Rochester:

Wonderful. Well, thank you both again, Dr. Gogoi, Dr. Gamble, thank you for this enlightening conversation. We’ve learned so much about endometrial cancer. I think for me, the main takeaway is postmenopausal bleeding, abnormal irregular bleeding is never normal. It needs to be evaluated. You both spoke about the importance of biopsy and not just an ultrasound, and in fact that ultrasound can be normal.

And with regard to disparities, I really appreciate both of your thoughts around community education, and this being a team sport and listening to our patients. So again, thank you so much for being here today, and thank you all for watching the program and tuning into this Empowering Providers to Empower Patients, Patient Empowerment Network program. I’m Dr. Nicole Rochester. Thanks again for watching.


Share Your Feedback

What Are AML Inhibitor Therapies and How Do They Work?

What Are AML Inhibitor Therapies and How Do They Work? from Patient Empowerment Network on Vimeo.

What are AML inhibitor therapies, and how do they work? Dr. Gail Roboz explains the different types of inhibitor therapies, their targets, the patient type they may benefit most, and a new class of targeted treatments.

Dr. Gail Roboz is director of the Clinical and Translational Leukemia Programs and professor of medicine at Weill Cornell Medicine and the New York Presbyterian Hospital. Learn more about Dr. Roboz.
 

 

Related Resources:

Choosing Therapy | How Are AML Treatment Goals Determined?

Choosing Therapy | How Are AML Treatment Goals Determined?

Expert Overview | AML Treatment Options and Phases of Therapy

Expert Overview | AML Treatment Options and Phases of Therapy

What AML Patients Should Understand About Test Results 

What AML Patients Should Understand About Test Results

Transcript: 

Katherine Banwell:

You mentioned one inhibitor as targeted therapy, but there are a couple of others. Would you briefly tell us about those? 

Dr. Gail Roboz:

So, over the years recently, we have identified certain specific targets in AML which are resulting in the addition of medications on these standard backbones. So, the target for venetoclax (Venclexta) is something called BCL2, and actually, venetoclax probably makes all chemotherapy better. It’s kind of a controversial statement, but I’m going to stand by it. But in AML, it has been shown that the addition of venetoclax to lots of different backbones makes them work better. There are other things to hit, though.  

For example, there are patients with AML who have something called a FLT3, F-L-T-3 mutation. This mutation also has specific inhibitors that are FDA-approved drugs that target specifically the FLT3 mutation, and if you have one of those, your doctor may add on a FLT3 inhibitor to either a lower intensity or an intensive backbone. Similarly, there are agents called IDH inhibitors. There are IDH1 and IDH2 inhibitors. 

If I start getting into isocitrate dehydrogenase pathways on this webinar, I think everybody will click off, because it’s certainly bored all of the medical students in med school, and it’s pretty tough to understand. But the bottom line is it’s very cool stuff because that boring pathway in medical school that nobody really thought about too much is actually part of very, very, central cellular functions that are a vulnerability now that have been identified in leukemic cells that, if you hit them with these specific inhibitors, patients do better.  

Now, couple of things for patients. It doesn’t mean that it’s better to have a FLT3 or an IDH mutation because the targeted therapies are available. So, a lot of patients are disappointed when they don’t have mutations. I don’t want you to think in that way. It’s not that it’s better, it’s different.  

It identifies a different biology. If you have certain mutations, there are certain medications that may help you more.  

That’s why I think the patients are learning quickly, too, to ask the doc – they may not remember the letters of the alphabet soup, but “Do I have something about my AML that can get one of these targeted therapies added on?” I think is a good question to think about. “Do I have something about my disease that has a specific drug that we’ve already learned makes outcomes better?”  

Katherine Banwell:

There’s a new emerging therapy as well. Is it the menin inhibitor? 

Dr. Gail Roboz:

I think that, in understanding different targets and different pathways, it leads me to a general statement that if you can get yourself potentially onto a clinical trial at an academic center, that is something to consider right out of the gate. Because there is a lot, a lot, a lot going on in this field right now. 

What we are hoping, and the reason that I am talking to you about venetoclax and FLT3 inhibitors and IDH inhibitors, is because of all the patients who jumped onto those clinical trials and proved that those drugs are better. Some of them are my patients! I was fortunate on some of those early trials to have some real winners in patients who got onto the trials. They’re the ones who drove the success. 

So, for example, menin inhibitors, which are very, very exciting, targeted agents for NPM1 and KMT2A mutations and rearrangements – these are complicated to remember as a patient, but there’s a cool drug out there that might be for you.  

I think that patients who really think about asking the question wherever they are, the “Hey, I just got a diagnosis of AML. Is there a clinical trial that might look good for me?” I think is a great question to ask pretty much out of the gate. 

Empowered Care: A Patient’s Guide to Navigating Endometrial Cancer

Empowered Care: A Patient’s Guide to Navigating Endometrial Cancer from Patient Empowerment Network on Vimeo.

Meet Sharon, a 61-year-old endometrial cancer survivor who knows firsthand the importance of being an active participant in her healthcare. After a two-year struggle with misdiagnosed symptoms and a dismissive doctor, she took charge and found the right medical support, leading to a diagnosis of endometrial cancer.

Sharon’s journey, from diagnosis to treatment and recovery, highlights the vital role of self-advocacy and patient activation. Her story is a beacon for others, especially women of color facing similar challenges, emphasizing the power of being informed, asking questions, and seeking support.

Download Guide | Descargar Guía en Español

See More from [ACT]IVATED Endometrial Cancer

Related Resources:

Addressing Disparities in Gynecologic Oncology | Key Challenges and Solutions

Addressing Disparities in Gynecologic Oncology | Key Challenges and Solutions

How Is Gynecological Cancer Care Impacted by Social Determinants of Health?

How Is Gynecological Cancer Care Impacted by Social Determinants of Health?

How Can Gynecologic Oncology Racial Disparities Be Addressed

How Can Gynecologic Oncology Racial Disparities Be Addressed

Transcript:

Being activated is a critical part of endometrial cancer care, especially for patients like me. 

My name is Sharon, I’m 61, and my diagnosis came after a two-year struggle with unidentified symptoms. Sharing my experience is my way of reaching out, hoping it might provide guidance to others facing similar challenges.

My symptoms began with abnormal vaginal bleeding, but my periods had ended over 10 years ago. That had me worried, and my sister encouraged me to see a doctor after I told her about the bleeding. I scheduled an appointment, but my doctor dismissed my symptoms. I felt like he wasn’t really listening to me and decided to find a doctor who looks more like me and would be more likely to listen to me. I found a Black female doctor who was concerned about my symptoms, which had worsened by then. I was also feeling pelvic pain. My new doctor scheduled an endometrial biopsy, and I was diagnosed with papillary serous carcinoma shortly afterward.

With an aggressive type of cancer, my oncologist scheduled a laparoscopic hysterectomy to remove my uterus along with my ovaries, fallopian tubes, and sentinel lymph nodes. I had the surgery within a few days, which was quickly followed by radiation to help ensure any remaining cancer cells were wiped out. My recovery went smoothly, and I continue to live a full life while getting regular scans to ensure I remain cancer-free.

After my cancer experience, I want to educate other women about what I’ve learned about endometrial cancer. Black women have nearly twice the death rate from endometrial cancer compared to white women. Hispanic, Black, and Asian women are not represented in clinical trials at equal rates to white women. And Black women are also diagnosed more frequently with rare but aggressive endometrial cancer forms. Remember that you shouldn’t have to suffer with your pain, and you can advocate for yourself and ask about patient advocates to advocate on your behalf.

Here are my activation tips for patients facing an endometrial cancer diagnosis:

  1. Ask your care team questions to learn about the status of your endometrial cancer, treatment options, and what to expect during and after treatment.
  2. Join a patient support group to offer and receive emotional support.
  3. Last but not least, inquire if a clinical trial may be a potential treatment option for your endometrial cancer.

Remember, stay activated by being informed, empowered, and engaged in your cancer care.


Share Your Feedback:

Create your own user feedback survey

Elevate | What You Should Know About Your Role in AML Treatment and Care Decisions Resource Guide

Download Resource Guide

PEN-208_ElevateAML_Guide_053024_F

Download Resource Guide

See More from ELEVATE AML

Elevate | What You Should Know About Your Role in AML Treatment and Care Decisions

Elevate | What You Should Know About Your Role in AML Treatment and Care Decisions from Patient Empowerment Network on Vimeo.

How can you elevate your AML care and treatment? AML expert Dr. Gail Roboz discusses the importance of participating in AML treatment decisions, reviews key factors that may impact therapy options, and shares advice for advocating for yourself.
 
Dr. Gail Roboz is director of the Clinical and Translational Leukemia Programs and professor of medicine at Weill Cornell Medicine and the New York Presbyterian Hospital. Learn more about Dr. Roboz.
 

Related Resources:

FLT3 inhibitors for AML Update

Thriving With AML | Advice for Setting Goals and Making Treatment Decisions

Thriving With AML | Advice for Setting Goals and Making Treatment Decisions

Expert Advice | Managing AML Symptoms and Treatment Side Effects

Expert Advice | Managing AML Symptoms and Treatment Side Effects

Transcript: 

Katherine Banwell:

Hello, and welcome. I’m your host, Katherine Banwell. It’s no secret that the quality-of-care patients receive can vary, and patients who are educated about their condition and involved in their care may have improved outcomes. That’s why the Patient Empowerment Network created the Elevate series, to help AML patients and their care partners feel well-informed when making treatment decisions with their healthcare team. 

In today’s program, an AML expert will join us to share advice for accessing better overall care. Before we get into the discussion, please remember that this program is not a substitute for seeking medical advice. Please refer to your healthcare team about what might be best for you. Well, let’s meet our guest today. Joining us is Dr. Gail Roboz. Dr. Roboz, would you please introduce yourself? 

Dr. Gail Roboz:

Absolutely. Thank you so much for having me. My name is Gail Roboz. I’m a professor of medicine and director of the clinical and translational leukemia programs at Weill Cornell Medicine and the NewYork-Presbyterian Hospital in New York City. Thank you again for having me. 

Katherine Banwell:

Well, thank you so much for joining us today. We really appreciate it. I’d like to start by discussing your role as a researcher. You’re on the frontlines for advancements in the AML field. What led you here, and why is it important to you? 

Dr. Gail Roboz:

So, I’m actually asked that question quite frequently, because AML is a challenging, difficult, scary disease, and people don’t necessarily assume that somebody in medical school would gravitate toward it. 

But I have to say that what is incredibly fascinating back then and now about leukemia is the continuous access to the disease. Patients will maybe giggle or groan as I’m saying that, because you can get a blood sample really anytime. You can even get a bone marrow sample anytime, although patients don’t enjoy that so much. 

But from a research perspective, it is absolutely extraordinary to be dealing with a disease where you can, in real time, truly run back and forth to a laboratory and see what’s happening, what is the new drug or the old drug doing, what’s happening with the patient, and I would say that from a fascination of a medical student perspective that grabbed me then and still does today.  

Katherine Banwell:

When it comes to choosing therapy for AML, it’s important to work with your healthcare team to identify what will work best for you, the patient. So, I’d like to know how you define shared decision-making.  

Dr. Gail Roboz:

The problem with AML sometimes is that it can be such an acute, emergency-type of presentation and urgent decision-making that I think your question is almost right out of the gate for some patients that will, “Wait, I don’t even have a minute, here. How do I build a team, do the research, look online if people are telling me that I’m in the middle of an emergency?”  

That isn’t always the case for acute leukemia, but it sometimes is. I think that what happens in AML in particular for patients is a building of knowledge and a building of the team, and figuring out, first of all, where am I when I am being told this diagnosis, and is it really an emergency? Do I have to make decisions really right now, because is it life-threatening today, I don’t have time to look around? Or do I have a minute to pause and get more information? 

I definitely feel that with the Internet era and with so much connection between doctors and teams, there is much more ability to reach out instantaneously for doctors, too, to get advice on a patient who might be in a smaller hospital that doesn’t have AML experience. But I think that the first thing is to try to figure out very, very quickly, what needs to happen to me as a patient immediately, and what can wait a minute, so that I can figure out what am I being told, and what are my options?  

Katherine Banwell:

Right, right. It can be confusing for patients, just finding out this new information. Part of making care decisions is setting goals. What are AML treatment goals, and how are they determined?   

Dr. Gail Roboz:

I would say that leaving cure on the table from the beginning is always a good place to start, because you want to figure out, first of all, what am I dealing with? What are the actual options?   

But when AML strikes, and a patient who has multiple medical conditions or comorbidities that are truly compromising function independently of the diagnosis of AML, that’s going to be a special path of what is actually reasonable for someone who is terribly medically ill or otherwise frail right from the beginning? That can be defining goals, but I think from the beginning, the best thing is to leave everything on the table. What can actually be done to make me better, first of all, to get me out of my immediate trouble? What can be done to make me better, and if I’m getting better, well, I like that, how do I stay there?  

What can be done to hang on to the state of ‘better,’ which is sometimes defined as remission? In AML, the goal is to get the bone marrow working again, functioning again, get rid of the acute emergency problem, if there is one, which there may or may not be in acute leukemia. 

Sometimes it’s truly an emergency, and sometimes it isn’t. But once I get better, can I stay there? What is required to keep me with a working bone marrow for as long as possible? 

But once you are starting to sort through the diagnosis, you realize that saying that somebody has acute myeloid leukemia is not telling me nearly enough information. This is a disease that is what we call biologically heterogeneous, which means there are lots of different forms. It’s like saying you’re sick. What exactly does that mean? There are lots of things that can make you sick. There are lots of different subtypes of AML, and fairly quickly in most institutions, we start getting back some information specifically on the subtype and biological characteristics of the disease.  

This can be very, very important in the initial treatment planning, and depending on where you are, the information that you get back can sometimes take 24 hours, 48 hours, 72 hours, a week. So, you start learning very quickly though that, “If I’m not in a complete emergency that requires instantaneous treatment, can I get back more information about the biological subtype of the disease so that I can start treatment planning of what is my best option right out of the gate?” That’s usually called induction, or the first therapy that you’re going to get with the goal, ‘getting rid of leukemia cells and getting into remission.’ That’s part one, and then everything that comes after that is about keeping you in remission.  

But for the initial goal, what is the therapy that the patient needs to get to get into remission? In order to figure that out, the good news is there are a lot of different ways to slice and dice getting into remission, and actually, it used to be such a weighty decision. 

Now, I would actually encourage people to – not relax, you can never use the word ‘relax’ with acute leukemia. But there are several different induction strategies for most patients that would be okay.  

So, even if you get started with one strategy and you hear five days later that another doctor might do something different, there are a lot of ways to safely get into remission. I think everybody should be pleased about the fact that we’re doing much better than we used to for patients across the board, all the way from children to much older adults, to safely getting people into remission. 

Katherine Banwell:

Right. So, what sort of factors then do you take into consideration when you’re choosing a therapy? 

Dr. Gail Roboz:

So, out of the gate, there are the patients that I think I referred to earlier who truly, truly are in situations based on their other diseases that there are certain treatments we would just cross out right out of the gate.  

If there are patients with very, very severely compromised cardiac or renal or lung function or are terribly ill from other conditions, AML doctors will right out of the gate for those patients eliminate certain treatments. But absent that scenario, what we try to look for is the biology of the disease. Not look at the age, not look at the comorbidities unless they are so severe that they make obvious certain choices. 

But rather, what I like to do is say, “What kind of AML is this, and what is the best treatment that I have to get this patient into remission?” And then ask the question, “can this particular patient handle this therapy?” Sometimes, these days, there actually may be more than one route to get to remission depending on the biology of the disease, and then, if that’s the case, then I can start getting picky and look at the individual patient. Where does the patient live? Who’s the patient’s family? What other diseases has the patient been treated for?  

Is there something that I can use? If I have a choice, if there are a couple of different things that might work, how do I fit the treatment to best take care of the needs of this particular patient? If I don’t have choices, then my question is, “Okay, how do I get this patient through my one therapy that I think is the truly, truly best option?” 

Katherine Banwell:

Okay. I’d like to turn to test results for a moment. What sort of tests should be done following an AML diagnosis?  

Dr. Gail Roboz:

We often generally recommend a bone marrow biopsy, even if we know we can make the diagnosis from a blood test, because even though the bone marrow biopsy is not the most fun test in the world, it does offer better information for follow-up care than what you can get initially from the blood. 

So, every once in a while, we do have a patient for whom a bone marrow biopsy itself for whatever reason can’t be done. But almost always, we need a bone marrow biopsy, and on that biopsy, you’re going to look under the microscope and see what the cells look like. You’re going to get back standard testing, which is called flow cytometry, which is going to tell the difference between what are the different cells that you’re seeing under the microscope. 

But then you’re actually going to get progressively much more fancy testing, including things called chromosomes or cytogenetics, and then ultimately, the majority of patients, if at all possible, will be having mutational testing to identify certain subgroups of AML that benefit from very particular treatments. Next-generation sequencing, PCR, fusion proteins, FISH, cytogenetics, I can go on and on with all kinds of terminology that is very confusing, even to hematology fellows, let alone to patients.  

Usually, we use a combination of tests to decide, “Is this patient likely to be able to be cured with chemotherapy alone, or might this patient benefit from a stem cell transplant from somebody else after they go into remission?” 

That’s basically what the prognostic scoring systems used to be asking, but now it’s a lot more complicated than that. Because even in the favorable categories, even in the adverse categories, where there used to be very little subtlety, now there is a lot of subtlety. 

It’s all about defining getting into remission, and what do I give you once you’re in remission to keep you there? It’s no longer this windshield wiper thing of good, bad, transplant, no transplant. There’s a lot more to AML than there used to be. 

Katherine Banwell:

I’d like to add that if you, the viewer, are interested in learning more about AML testing and treatment, PEN has a number of resources available for you. You can find these at powerfulpatients.org/AML, or by scanning the QR code on your screen.  

Before we get into specific treatment types, Dr. Roboz, would you provide a brief explanation of the phases of therapy for AML? You mentioned induction therapy earlier. Would you tell us what that is? 

Dr. Gail Roboz:

Yeah. So, here, too, I have to say that it’s more confusing than it used to be for the following reasons. So, historically and currently, we typically talk about induction as the first therapy that you’re going to get to get into remission.  

Then, the treatment paradigm is you do something to get into remission; do some treatment to get into remission. After that, in the realm of post-remission therapy, there are different things that can happen. There can be something called consolidation, which might be another round of chemotherapy. Some patients get consolidation, some patients don’t. After consolidation, there can be a transplant.  

So, you get into remission, you may or may not get a little bit of what’s called consolidation chemotherapy, and then go on to a transplant. 

However, sometimes either after the transplant or after chemotherapy before ever getting or instead of ever getting a transplant, there might be ongoing treatment in a lower intensity ongoing basis that is called maintenance.  

So, you’ve got to think about it as induction as what happens first, consolidation is something that happens when you’re in remission, and then maintenance usually refers to ongoing therapy that is different from consolidation. 

It’s usually lower intensity, easier to take, oral types of treatment that may go on and on. And just to be incredibly confusing, it’s different from something like breast cancer, where often the patients are given, “You get six cycles of this, and then you’re done.” From AML, there’s actually often not that type of an obvious plan right out of the gate for the patient. 

The answer will be, “It depends.” It depends. It depends how your treatment looks at this point in time. It depends how you look at this point in time. 

So then, the patients say, “Well, aren’t you going to cure me of this? What are you doing? Aren’t you going to get rid of it?” So, historically, there are some patients who get cured with chemotherapy. They get chemotherapy to get into remission, they get some chemotherapy afterwards, and there’s a cure rate for some patients with that. The majority of patients who are cured with AML get an allotransplant, or a transplant from somebody else. 

Then there’s a whole group of patients where we’re asking the question now, is it possible to get those patients beyond five years – so in oncology, five years is typically defined as cure. Can we get some patients with ongoing therapy to that past-five-year mark without a transplant? That’s in the zone of the ‘coming soon.’ Don’t have a ton of patients in that group right now, but hopefully we will. 

Katherine Banwell:

You’ve mentioned some various treatment types that are used to treat AML. Can you share a brief overview of available treatments? 

Dr. Gail Roboz:

So, the terminology that we use is a little bit annoying, because it is a little bit general. We say intensive and not intensive. 

But historically, intensive chemotherapy referred to a combination of generally two types of agents, cytarabine (Cytosar-U) and an anthracycline, which is a class of chemotherapy, that either just those two together or in combination with sometimes a third or a fourth drug usually keeps people in the hospital for around a month. Not that the chemotherapy takes that long, but the treatment gets rid of basically a lot of cells in the bone marrow, good guys and bad guys, and it takes about three weeks for those normal cells to recover. 

So, a standard intensive induction for AML is often around three to four weeks in the hospital, somewhere between three and five or so days of chemotherapy up front, depending on exactly what the protocol is. The classic regimen is actually still called 3+7, three days of one drug, seven of the other. But there are many variations of that that work. 

The chemo is then stopped, the patient hangs out in the hospital, very frequently getting transfusions and antibiotics, and we wait for the bone marrow to recover.  

Another current path that many patients are getting – almost all older patients, with ‘older’ being defined not by a specific age cutoff, but often 75 and older, almost everybody agrees no longer gets the classic chemotherapy that I just described. At some institutions, that 75 is going down, and even 70 and 65 and above are getting a new type of therapy, mostly because the new type of therapy is working pretty well. That is a combination of something called a hypomethylating agent.  

Drugs like azacitidine (Vidaza, Onureg) or decitabine (Dacogen) in combination with a pill that has changed the landscape of AML more than any other called venetoclax (Venclexta). Venetoclax is a drug that is not exclusively used for AML. 

It actually was originally approved for another type of leukemia. But I think that not many people would argue with the statement that what has changed absolutely the face of AML treatment has been this drug, because it’s a BCL2 inhibitor. What it does is it actually – cancer cells and leukemia cells in particular are very, very good at staying alive.  

They don’t undergo cell death, they don’t want to die, and venetoclax brings down their forcefield so that those cells can actually undergo apoptosis and die. 

Venetoclax in combination with azacitidine or decitabine has transformed the care of the disease, because many patients older than 65 – and the median age of diagnosis of AML is around 68 to 70. So, many patients never were well enough to have the intensive therapy. They weren’t going into remission, and they weren’t having prolonged survival often beyond a few months. 

But now, those patients do actually much better with the combination of aza [azacitidine] and venetoclax. So typically, the induction path is going to be deciding who gets an intensive therapy backbone, usually associated with long hospitalization. Who gets a less intensive backbone – by the way, that is often associated with just the same hospitalization. So, that’s why I don’t love the term ‘low intensity,’ because that implies that it doesn’t work.  

It does, and it also implies that you’re not going to be in the hospital. You probably will, because in the same way as for the more so-called intensive therapies, getting into remission involves getting rid of bone marrow cells and waiting for the normal ones to recover. Even if you are a patient who is getting the venetoclax combined with the azacitidine or decitabine, which is typically called low intensity, you may very well be in the hospital for a month. 

Because depending on where you live and who your family is and how sick you might be, you will probably want us to watch you carefully during that first month, but it’s worth it. Because if you have a good chance of getting into remission, remission is what makes life better and life longer. So, we want to get patients into remission, even if it means upfront time in the hospital. 

Katherine Banwell:

You mentioned one inhibitor as targeted therapy, but there are a couple of others. Would you briefly tell us about those? 

Dr. Gail Roboz:

So, over the years recently, we have identified certain specific targets in AML which are resulting in the addition of medications on these standard backbones. So, the target for venetoclax is something called BCL2, and actually, venetoclax probably makes all chemotherapy better. It’s kind of a controversial statement, but I’m going to stand by it. But in AML, it has been shown that the addition of venetoclax to lots of different backbones makes them work better. There are other things to hit, though.  

For example, there are patients with AML who have something called a FLT3, F-L-T-3 mutation. This mutation also has specific inhibitors that are FDA-approved drugs that target specifically the FLT3 mutation, and if you have one of those, your doctor may add on a FLT3 inhibitor to either a lower intensity or an intensive backbone. Similarly, there are agents called IDH inhibitors. There are IDH1 and IDH2 inhibitors. 

If I start getting into isocitrate dehydrogenase pathways on this webinar, I think everybody will click off, because it’s certainly bored all of the medical students in med school, and it’s pretty tough to understand. But the bottom line is it’s very cool stuff because that boring pathway in medical school that nobody really thought about too much is actually part of very, very, central cellular functions that are a vulnerability now that have been identified in leukemic cells that, if you hit them with these specific inhibitors, patients do better.  

Now, couple of things for patients. It doesn’t mean that it’s better to have a FLT3 or an IDH mutation because the targeted therapies are available. So, a lot of patients are disappointed when they don’t have mutations. I don’t want you to think in that way. It’s not that it’s better, it’s different.  

It identifies a different biology. If you have certain mutations, there are certain medications that may help you more.  

That’s why I think the patients are learning quickly, too, to ask the doc – they may not remember the letters of the alphabet soup, but “Do I have something about my AML that can get one of these targeted therapies added on?” I think is a good question to think about. “Do I have something about my disease that has a specific drug that we’ve already learned makes outcomes better?”  

Katherine Banwell:

There’s a new emerging therapy as well. Is it the menin inhibitor? 

Dr. Gail Roboz:

I think that, in understanding different targets and different pathways, it leads me to a general statement that if you can get yourself potentially onto a clinical trial at an academic center, that is something to consider right out of the gate. Because there is a lot, a lot, a lot going on in this field right now. 

What we are hoping, and the reason that I am talking to you about venetoclax and FLT3 inhibitors and IDH inhibitors, is because of all the patients who jumped onto those clinical trials and proved that those drugs are better. Some of them are my patients! I was fortunate on some of those early trials to have some real winners in patients who got onto the trials. They’re the ones who drove the success. 

So, for example, menin inhibitors, which are very, very exciting, targeted agents for NPM1 and KMT2A mutations and rearrangements – these are complicated to remember as a patient, but there’s a cool drug out there that might be for you. I think that patients who really think about asking the question wherever they are, the “Hey, I just got a diagnosis of AML. Is there a clinical trial that might look good for me?” I think is a great question to ask pretty much out of the gate. 

Katherine Banwell:

The symptoms of AML as well as the side effects of certain medications can vary greatly among patients. So, how do you approach symptom management with your patients? 

Dr. Gail Roboz:

Patients will giggle because I repeat this line. You have to be afraid of the disease, not the treatment. I think that if you read the package insert on a Tylenol, you’re certainly not going  to think you’re going to live for more than 20 minutes if you take one of those. You can certainly appreciate that, with chemotherapy drugs and including some of the novel agents that I’m talking about, if you read package inserts and look at some of the signs and symptoms and things that can happen, it’s extraordinarily overwhelming. 

I think that a lot of what I do for patients is I keep them close. Because if the patient is in the hospital or coming in very frequently in clinic, I think that that everyday assessment of, “What are you experiencing?” and “What can I tell you is the disease’s fault, and what can I tell you is the medication’s fault?” is so, so important. 

Especially in the newly diagnosed patients, where the disease is active. Of course, we want to try to minimize anything that we can do to make the process better for patients, more comfortable for patients, but there are certain things that we do tell people, “You’ve got to slug through this particular problem, because this is the disease’s fault.” This is different from a patient in remission, where they might be getting ongoing therapy with something, or we say, “Hey, wait a minute. You’d be feeling fine, except now you’re taking this medication. How do we minimize messing up quality of life in remission?” 

Because we want you to feel great when you’re in remission. I think the real answer of that is to have a really close collaboration with the healthcare team, and for the patients to really understand – I repeat this because it’s so important. What is the disease’s fault, and what is the treatment’s fault? If there’s something that is therapy-related, do I have a substitute or do I not have a substitute?  

Because if the drug is essential to get us where we need to go, well, what can we do to manage comfort and to manage symptoms until you get to the place where your marrow is working again? 

Katherine Banwell:

That’s great advice, Dr. Roboz. I would like to get to an audience question that we received prior to the program. This one comes from Johanna. “How can I better understand my lab test results? What questions should I be asking my provider about those results?”  

Dr. Gail Roboz:

One of the things that I would say to patients is to be careful when interpreting your own results, because I really am not exaggerating to say that patients have had absolute trauma looking at things that I look at it and say, “Oh, this looks great.” So, the first thing is, be careful being your own doctor. 

The second thing though is that the author of the question has to understand that there’s going to be a tsunami of data coming in with respect to AML treatment. Sometimes in the hospital on a daily basis when you’re in the middle of an induction, there is a true – tsunami is the right word – a deluge of data, and you have to work with your team to say, “What am I following here? What’s important at this phase in my treatment? What’s the number I’m looking at?” Patients sometimes tell me, “I don’t want to know any of this,” and I’m fine with that.  

I think it’s actually okay. Sometimes patients will say, “Give me guidance,” and I will be specific. Because you can actually have a leukemia induction patient where every single laboratory value is abnormal. They might be getting pushed to a device, in the morning, sitting in the hospital on your iPad, 50 abnormal results. You’re trying to battle back the disease and be positive and advocate for yourself, but there are 50 abnormal results in front of you. 

I think you have to really work with the team to say, “What am I looking at today? What are the numbers that are the really important ones? There are 50 abnormal ones here; everything is getting a yellow or a red light in this. How do I go through this?”  

And to appreciate, also, that at different points in the treatment, the beginning of treatment induction post-remission therapy, you’re looking for different things. So, work with your team so that you’re not assessing every single result with equivalent weight, because I think you’re going to stress yourself out.  

Katherine Banwell:

That’s great advice, Dr. Roboz. Thank you. As we close out the program, I’d like to find out what you would like to leave the audience with. Why are you hopeful? 

Dr. Gail Roboz:

AML is changing incredibly rapidly. And  I can tell you it is a lot more fun to be an AML doctor now than it used to be, with respect to what I am offering for patients. We have always fought really, really hard to have our wins, but we’re winning more. I do think that it is a complicated space to navigate for patients, but there is room for a lot of optimism. 

I think we are getting patients transplanted  –  patients that we never thought would ever go through a transplant or getting transplanted. Patients who never had a chance of even living more than six or eight months or living much longer than that. Is it perfect? No. Do we have as many cures as we want?  

No, but there’s a lot going on. I think if patients feel that excitement, they will also feel the need to ask about those clinical trials. Because I think that for a lot of patients, clinical trials is an area where they would be worried. They’re not sure that they want to. “I don’t want to be a guinea pig,” and yet here I can say in the AML space, one after another after another drug approvals in the last several years, with the patients on those trials being awfully happy that they participated. 

So, I think that it’s a very, very terrifying diagnosis. There’s nothing that I can do to take the sting out of that. But try to find yourself in an optimistic place with options that are being offered to the very, very, very best that we can do. There are patients who are listening, I’m sure, who have relapsed or refractory disease who are not feeling that optimism. 

I want to address you specifically, because we don’t have enough yet. We’re trying. When you have AML that has come back or come back multiply, that’s dangerous and difficult. But for those patients in particular, try really hard to get onto clinical trials. If the drugs that we have out there – if you’ve already taken them and they haven’t worked for you or if they’re not serving you well, if you’re in good shape and the drugs that we have aren’t good enough, well, let’s see if we can get you on something that’s investigational. 

Katherine Banwell:

Dr. Roboz, thank you so much for taking the time to join us today. 

Dr. Gail Roboz:

Thank you for having me. 

Katherine Banwell:

I also want to thank all of our collaborators. To learn more about AML and to access tools to help you become a proactive patient, visit powerfulpatients.org. I’m Katherine Banwell. Thanks for joining us today.  

What AML Clinical Trial Inequities Do Minority and Ethnic Groups Face?

What AML Clinical Trial Inequities Do Minority and Ethnic Groups Face? from Patient Empowerment Network on Vimeo.

Do minority and ethnic groups face acute myeloid leukemia (AML) clinical trial inequities? Expert Dr. Andrew Hantel from Dana-Farber Cancer Institute and Harvard Medical School discusses research study results of NCI-designated cancer centers of AML versus other cancers and proactive patient advice for clinical trial access.

[ACT]IVATION TIP

“…patients to ask about where the nearest sites of care are for them that have clinical trials on at least historically, and then reaching out to those sites to ask, ‘Can I have a consultation?” Am I somebody who might be eligible for any of the clinical trials that you have?’”

Download Resource Guide | Descargar guía de recursos

See More from [ACT]IVATED AML

Related Resources:

What Are Key Acute Myeloid Leukemia Care Barriers and Solutions?

What Are Key Acute Myeloid Leukemia Care Barriers and Solutions?

Why Is Post-Access Enrollment Vital in AML Clinical Trial Participation?

Why Is Post-Access Enrollment Vital in AML Clinical Trial Participation?

Underrepresented AML Clinical Trial Groups | What Solutions Are Underway

Underrepresented AML Clinical Trial Groups | What Solutions Are Underway

Transcript: 

Lisa Hatfield:

Dr. Hantel, what primary findings are in your study regarding inequities and clinical trial participation among minoritized race and ethnic groups with cancer at the comprehensive cancer centers?

Dr. Andrew Hantel:

So we performed a couple of different studies on this, I think together, they’ve collectively found that there are significant disparities in clinical trial participation among minoritized racial and ethnic groups at comprehensive cancer centers. And just to take a step back and say, what are comprehensive cancer centers? They’re basically a designated center, and the designation is provided by the National Cancer Institute (NCI), and that basically says that this place is of such and such a level of quality in terms of their delivery of cancer care and their research that they perform, and so these are generally larger academic centers that have a lot of clinical trials, have a lot of experts across different cancer types, including AML. And those are the ones that we are wondering if people had equal or equitable access to, in other cancers compared to AML.

A lot of the disparities in clinical trial participation is really because these different minoritized groups have less access to these comprehensive cancer centers, so they could really never be considered for trials, because they’re just not seen at places where the trials are taking place. This is the case for a lot of common solid tumors, such as breast and colon cancer, where very, very large proportions of patients are seen in the community.

And this means that in leukemia, however, we found that participation disparities were not only due to access, but because the leukemia is less common because a lot of docs actually almost preemptively refer some of their patients to get seen, some of the disparity is kind of shifted, and it ends up becoming not as much an issue just of access but also of getting into the trial after they’re seen at the center.

And this can be because of a variety of things, there are reasons that we can go into in a second, but we kind of also want to make the statement in the context of it not being a one-size-fits-all answer. There are some cases where comprehensive cancer centers actually both allow great access and allow equitable enrollment on their trials, and there are other comprehensive cancer centers that have had…and continued to have issues with this. 

So I also just want to make sure to make that distinction. But after somebody gets down to a center, you have the process of, is there a trial at the center that’s actually right for your specific type of leukemia and the stage of the disease, and if you’re up front or relapsed or all of these other nuances, and then you have the questions of, are you eligible, are you offered that trial, and then are you interested in partaking in that trial, and is it feasible for you to participate?

So there are all these other steps after that point of access, and in general, what we saw was that it was kind of these later steps that were as much more of an issue for equitable enrollment for leukemia as that first step of access. And so it’s slightly different from what we’ve seen in other cancers, and so the answers and the solutions for that are going to be different.

So my activation tip for this question would be for patients to ask about where the nearest sites of care are for them that have clinical trials on at least historically, and then reaching out to those sites to ask, “Can I have a consultation?” Am I somebody who might be eligible for any of the clinical trials that you have?” That might be something where it does take you to that site to actually get evaluated and see if there is anything available, but that would be the first step.

Share Your Feedback About [ACT]IVATED AML

What Are Key Acute Myeloid Leukemia Care Barriers and Solutions?

What Are Key Acute Myeloid Leukemia Care Barriers and Solutions? from Patient Empowerment Network on Vimeo.

What are key barriers and solutions to acute myeloid leukemia (AML) care? Expert Dr. Andrew Hantel from Dana-Farber Cancer Institute and Harvard Medical School discusses factors that create AML care barriers, solutions to overcome barriers, and proactive patient advice to help ensure optimal care.

[ACT]IVATION TIP

“…patients and their families to be aware of where any specialized care facilities are near them, it might not be even so much geographically near as like within the same 10 miles, but it might be like, ‘What’s the nearest large facility that maybe is known for having clinical trials or other medical research?’ And just to reach out to them and ask, ‘Do you have telehealth consultations? What would it mean for me to come to you?’…and then talk to the actual physician who’s caring for them and say, ‘Is it safe for me to wait to talk to them, is it something where I do therapy now, and I should maybe go talk to them after I’ve received my initial therapy and have that discussion?’”

Download Resource Guide | Descargar guía de recursos

See More from [ACT]IVATED AML

Related Resources:

What AML Clinical Trial Inequities Do Minority and Ethnic Groups Face?

What AML Clinical Trial Inequities Do Minority and Ethnic Groups Face?

Why Is Post-Access Enrollment Vital in AML Clinical Trial Participation?

Why Is Post-Access Enrollment Vital in AML Clinical Trial Participation?

Underrepresented AML Clinical Trial Groups | What Solutions Are Underway

Underrepresented AML Clinical Trial Groups | What Solutions Are Underway

Transcript: 

Lisa Hatfield:

Dr. Hantel, what are some of the key barriers that patients with AML encounter when accessing timely and effective care, and how can healthcare systems work to address these challenges?

Dr. Andrew Hantel:

That’s a great question. People with AML face significant barriers in accessing care, primarily due to the complexity and intensity of the treatment options that we have available. The landscape of those treatment options is really much different than it was even 10 years ago. We’ve had more than 10 novel drug approvals and extensions of treatments that are effective into groups such as older adults who have really previously had few good options and now have potentially many, and this has led to differences in what some of us call diffusion of innovation or how treatments are taken up and used by the medical community.

We kind of have some people who are usually at academic centers and are early adopters because they’ve seen some of these newer drugs and participate in the global trials, and then we have other community docs who see a lot of people with a variety of different cancers and kind of keep up with the literature, but to some extent aren’t as familiar with those new drugs and take some time before they start to incorporate them into practice.

And it is really important because in AML is not a disease that’s so common, so every oncologist sees patients with AML very frequently, and that kind of creates a difference between patients, people who are specialists, who are at the larger academic centers and people who are on the community and their need to really work together.

And this is a big point because treatments generally need to occur within a few weeks of diagnosis, if not much sooner, and therefore, all together this idea of new treatments kind of expanding out into the universe and also having the need for quick treatment means that geography really plays a crucial role in acting as a barrier to some patients getting what we think of as optimal care, especially for patients who live in rural or underserved areas that might not have access to these larger medical centers with specialized physicians, and then we also have financial challenges for those same patients and that getting into places is a major barrier, insurance to get to those places may have additional burdens that are placed in people with high costs that are associated with some of these treatments and healthcare systems have begun to address some of these challenges.

By really trying to enhance some local capacities such as through telemedicine consultations, kind of working in what we consider like a hub and spoke system where they have local providers reaching out to them and patients coming to them, at the time point of treatment decisions and then going back locally for a lot of their care, just so it’s not as burdensome in terms of travel, and then there’s also a lot of programs that are both within medical systems and outside of medical systems that are being helpful for patients, for financial guidance and assistance, such as through Leukemia & Lymphoma Society, which has wonderful programs, as well as kind of identifying partners for travel and housing grants and stipends that might be needed for caregivers, a lot of.

As I said, in a lot of centers like ours who are really trying to use any means possible to overcome these barriers for different patient groups, and a lot of it really depends on exactly what the person’s situation is, because so much of leukemia care is about the values of the patient and really how we’re targeting treatment, not only toward their disease, but really aligns with their goals, and so overcoming barriers is a really personal thing based on the values and the goals of the person who is in front of you.

Lisa Hatfield:

Great, thank you. And then one follow-up question I did have to that, as you mentioned that some patients are seen by their community oncologist, is it possible for a patient who is diagnosed with AML to do maybe one consult via telemedicine with a specialist in AML and maybe coordinate care that way, or do specialists typically like to have those patients present for regular visits?

Dr. Andrew Hantel:

Yeah, so I think the landscape of this is shifting in the past, I think no matter what, we have the idea that patients need to be safe, especially in the early days of their diagnosis, so it might be the case that somebody’s leukemia is diagnosed and they need treatment, it started immediately, and it’s not safe for them to wait, and it’s not safe for them to travel. But definitely there are other people who see their doctor and their blood counts are relatively stable, and the doctor thinks that it’s reasonable for them to get a second opinion, and these days, those opinions are happening yet still in person, sometimes centers like to see people in person.

But other times, as we said, those travel and distance and burdens can be so much that patients and these centers are turning to telehealth and other virtual forms of care that can at least provide a preliminary consultation and say, if you were to be seen here, we may have additional options like clinical trials or from what I’ve seen, I agree with what your physician is offering you and those more higher level decisions. And so it’s not so much whether or not even telehealth versus nothing, it’s kind of telehealth in person versus the need to get care immediately.

Lisa Hatfield:

Okay, that’s super helpful, thank you. And do you have an activation tip for this question, Dr. Hantel?

Dr. Andrew Hantel:

Yeah, I think the first thing is essentially just for patients and their families to be aware of where any specialized care facilities are near them, it might not be even so much geographically near as like within the same 10 miles, but it might be like, “What’s the nearest large facility that maybe is known for having clinical trials or other medical research?” And just to reach out to them and ask, “Do you have telehealth consultations? What would it mean for me to come to you?”

Those kinds of things, and then talk to the actual physician who’s caring for them and say, “Is it safe for me to wait to talk to them, is it something where I do therapy now, and I should maybe go talk to them after I’ve received my initial therapy and have that discussion?” And I think that leads into my kind of second or ancillary tip is to really don’t be afraid to ask your care teams about both of those things, and then about the logistics of what any of your care is going to mean in terms of the burdens of getting back and forth to clinic, having to be in the hospital and both for you as a patient as well as for your family.

Share Your Feedback About [ACT]IVATED AML

What Are the Advantages of Seeking Care With a Lung Cancer Specialist?

What Are the Advantages of Seeking Care With a Lung Cancer Specialist? from Patient Empowerment Network on Vimeo.

What are the benefits of seeing a lung cancer specialist? Dr. Thomas Marron discusses the key advantages of specialty care, the value of a second opinion, and options for seeing a lung cancer specialist via telemedicine.

Dr. Thomas Marron is Director of the Early Phase Trials Unit at the Tisch Cancer Institute at Mount Sinai Hospital. Dr. Marron is also Professor of Medicine and Professor of Immunology and Immunotherapy at the Icahn School of Medicine at Mount Sinai. Learn more about Dr. Marron.

See More from EVOLVE Lung Cancer

Related Resources

Advances in Targeted Lung Cancer Treatments | What You Should Know

Advances in Targeted Lung Cancer Treatments | What You Should Know

Antibody Drug Conjugates for Lung Cancer | Advances in Research

Antibody Drug Conjugates for Lung Cancer | Advances in Research

Lung Cancer Treatment Plan Advice | Where Do Clinical Trials Fit In?

Lung Cancer Treatment Plan Advice | Where Do Clinical Trials Fit In?

Transcript:

Katherine Banwell:

What’s the advantage then of seeking care with a lung cancer specialist? 

Dr. Thomas Marron:

So, I think it’s extremely important. Unfortunately, a lot of the country, there are not lung cancer specialists available around the corner. But in large cities, there’s typically many lung cancer specialists, but I think it’s extremely important, at least as a second opinion, even if you’re not going to be treated locally by a lung cancer specialist, to seek out expertise.  

And often times, I’ll have patients that come from more rural areas outside of New York and they’ll come, and they’ll see me and then I’ll work with their local provider to come up with a treatment plan. Because the fact of the matter is, is that in every cancer type, but particularly in lung cancer, the field is moving so quickly. So, the treatment options that we have available today were not available in 2022.  

And we’re going to have probably five to 10 drugs that’re going  to be FDA-approved in the next year. And it’s typically the lung cancer specialist where it’s all that we do, we eat, breathe and live lung cancer, we’re the ones that really are up to date on everything. While if you’re seeing a general hematology, oncology provider who I’m always in awe of, they have to stay up to date on lung cancer, breast cancer, lymphoma, leukemia, everything under the sun.  

And when you have so much development in the research that’s happening, you really want to be talking to somebody, at least as a second opinion that knows exactly what the most latest data is and what the best options are available. And also, those lung cancer providers are usually the ones that will know exactly where you can go to get access to certain clinical trials.  

Katherine Banwell:

In seeking a second opinion, can somebody do a tele-visit, or do you have to actually, physically go to see the specialist? 

Dr. Thomas Marron:

So, it depends on the specialist that you’re trying to see.  

There are certain institutions that will allow you to do televisits. Oftentimes doctors, at least for their first encounter with a patient really want to see somebody in person, just so that we can really evaluate how functional somebody is. There’s a lot that I cannot tell through my computer screen, through my Zoom call with a patient.

And so, it can be a little bit difficult, but there are many centers, including our own that will offer patients televisits as a second opinion, for us to get a chance to talk to them about their medical history, review, the treatment decisions that they’ve had in the past or the current treatment decision that they’re dealing with and give our own opinion on what they should do.  

Lung Cancer Treatment Plan Advice | Where Do Clinical Trials Fit In?

Lung Cancer Treatment Plan Advice | Where Do Clinical Trials Fit In? from Patient Empowerment Network on Vimeo.

What questions should patients ask about a lung cancer treatment plan? Lung cancer expert Dr. Thomas Marron shares key considerations when choosing therapy and discusses where clinical trials fit into planning.

Dr. Thomas Marron is Director of the Early Phase Trials Unit at the Tisch Cancer Institute at Mount Sinai Hospital. Dr. Marron is also Professor of Medicine and Professor of Immunology and Immunotherapy at the Icahn School of Medicine at Mount Sinai. Learn more about Dr. Marron.

See More from EVOLVE Lung Cancer

Related Resources

Advances in Targeted Lung Cancer Treatments | What You Should Know

Advances in Targeted Lung Cancer Treatments | What You Should Know

Expert Perspective | New and Emerging Progress in Lung Cancer Treatment

Expert Perspective | New and Emerging Progress in Lung Cancer Treatment

What Are the Advantages of Seeking Care With a Lung Cancer Specialist?

What Are the Advantages of Seeking Care With a Lung Cancer Specialist?

Transcript:

Katherine Banwell:

What questions should patients be asking about their proposed treatment plan? 

Dr. Thomas Marron:

I think that in lung cancer, most patients are going  to get the same therapeutic approach offered to them wherever they go.  

It’s not like certain types of cancer where there’s 10 different ways to treat it. But there are some nuances and depending on the location in which you’re getting treated, whether it be in an academic hospital or a community setting, you may have different chemotherapies offered, immunotherapies offered. You may have different combinations offered. And so, I think it’s important to always ask your provider what other options are there, and why are they recommending one option over another. But I think it’s also really important that patients get second opinions.   

A lot of my patients, even my in-laws are always very skittish about getting a second opinion because they don’t want to insult their doctor, who they feel very close to. And I would say, it couldn’t be further from the truth. Any good doctor is 100 percent okay with a patient going and getting a second, third, fourth opinion because to us, the most important thing is that you have confidence in the decisions that we’re making about your treatment.

I always tell patients, I’m basically a waiter here offering you a menu of options and giving you my recommendation. But it is up to the patient in the end what treatment they receive and how long they receive it for.  

And if they decide ever to discontinue it. And I think that the more information, the more smart people looking at you, the better.  

Katherine Banwell:

Where do clinical trials fit into a non-small cell lung cancer treatment plan? 

Dr. Thomas Marron:

So, that’s a phenomenal question and one that I hope that everyone asks their providers when they see them because the reality is that while we are curing some patients, the vast majority of patients are not cured. And I think that all patients should at least consider a clinical trial, whether it be a first line clinical trial. So, the first medicine that you receive for your cancer, or at the time of progression.  

I think particularly, once patients progress on the first line therapy, those patients we really don’t have a cure for, even if we have some palliative chemotherapies or eventually these antibody drug conjugates to treat them.  

And so, I think everybody who is progressing on first line therapy should always consider a clinical trial. And I think it’s extremely important that patients realize the need to ask their providers about clinical trials, but also be an advocate for themselves and go out and get second opinions, get third opinions and see what trials are available in the community and even in other cities.

Because often times in New York City, I’ll have completely different clinical trials than my colleagues at the other five institutions in the city. And it’s really important that patients advocate for themselves, and they identify everything that’s available.   

Newly Diagnosed Non-Small Cell Lung Cancer | Key Advice for Patients

Newly Diagnosed Non-Small Cell Lung Cancer | Key Advice for Patients from Patient Empowerment Network on Vimeo.

What’s key advice for newly diagnosed non-small cell lung cancer (NSCLC) patients? Expert Dr. Christina Baik from Fred Hutchinson Cancer Center discusses genetic testing, essentials to know about your lung cancer, and patient tips to ensure your best care.

Download Resource Guide

See More from START HERE Lung Cancer

Related Resources:

Lung Cancer Patient Expert Q&A: Dr. Christina Baik

Lung Cancer Patient Expert Q&A: Dr. Christina Baik 

Bridging gap

Advancing Lung Cancer Treatment: Bridging the Gap in Personalized Care

DNA

Understanding Oncogene-Driven Lung Cancer: Targeted Therapy Advances and Challenges 

Transcript:

Lisa Hatfield:

When a patient is diagnosed with non-small cell lung cancer, is genetic testing always done on the tumor, or do patients know what their mutations are right upon diagnosis if testing is done?

Dr. Christina Baik:

So, as a rule of thumb, they should, all patients should be tested, and there are exceptions. So, for example, in lung cancer, there’s the type that we call small cell lung cancer, and there’s non-small cell lung cancer. So we often, we usually do not do genetic testing on small cell lung cancer, because often these tumors do not have a genetic abnormality that for which we can actually give treatment for. But for non-small cell patients, I would say, if most, my personal opinion is that everybody should be tested with the genetic test and really advocate for that. You know, there are certain types of non-small cell lung cancer where there are genetic targets that are rare, however, you don’t know unless you test. So I would say yes to that question of testing for genetic abnormalities.

Lisa Hatfield:

Okay, thank you. So can you speak to the priorities for newly diagnosed patients, particularly populations who may have poor outcomes?

Dr. Christina Baik:

So, I think there are priorities when it comes to research, and then there are priorities for individual patients, right? So from a research standpoint, as I mentioned before, I think really the priorities, the priority is to develop strategies so that we’re truly personalizing treatment for each patient, and we’re not giving this kind of generic treatment for a bulk of the patients. So from a research standpoint, really understanding the biology, understanding what works for what patient, I think that’s extremely important.

On the individual patient level, we sort of alluded to this earlier, but really knowing the cancer we’re dealing with is extremely important. Know your cancer stage, ask what your cancer stage is, know the type of lung cancer that you have. So I will say as of now, there are, I can think of 12 or 13 different types of lung cancer that I want to make sure I know that patients, you know, what their subtype is.

So know your subtype of lung cancer. Ask those questions. If the knowledge is not known, if they say, “You know your stage is not very clear, your subtype is not clear,” then ask why that is, what type of additional testing that needs to be done. So I think those are the type of questions that each patient and their family member should really ask. And in terms of the poor outcome question, I think the first thing I would say is if a doctor tells you, you belong to a group of patients who are going to have a poor prognosis, then ask why that is, right? And understand the reasons for that.

And if that’s, once you understand, I think I’m a big proponent of getting second opinions, because a lot of these treatments and there’s a lot of medical judgment involved when we recommend treatments, and you just want to get a different perspective with the same type or set of information. So really being an advocate for yourself, I think that’s extremely important when you’re first diagnosed.

Lisa Hatfield:

Great, thank you. You mentioned two things I also feel strongly about, I don’t have lung cancer, I have a different type of cancer, but you said that patients and family members can ask questions.  Having an advocate with you at all times, if that’s possible, a family member, a friend going with you, I think is super important.

And also getting a second consult to understand your diagnosis better. I appreciate you saying that, because some of us are a little bit reluctant to do that, maybe afraid of offending our doctors. So, I appreciate that as a patient myself, so thank you. Okay. So talking about disease progression and recurrence, particularly for metastatic non-small cell lung cancer, what should patients know?

Dr. Christina Baik:

Okay. So when a cancer initially responds to a treatment and it stops responding, there can be many reasons for that. So the first question to really think about is is there another test we can do to identify the reason for the progression? And can we personalize a treatment according to that resistance pattern or the change that occurs in the tumor? This is more relevant to patients who get a targeted therapy, but I think it’s a good sort of rule of thumb in terms of asking your doctor why that is, and is there more testing that’s required?

And the second I would say is once the cancer progresses after the initial treatment, then, unfortunately, in lung cancer the treatment options are much more limited, and the effectiveness is very limited as well. So, it’s really at that juncture to really seek out clinical trials. There are many trials that are out there. So really working with your doctor in identifying these trials. If there is an academic center that’s close to you, at least inquiring about that. In lung cancer, fortunately, there are many wonderful advocacy groups and these advocacy groups can be great resources in finding out about clinical trials and where to seek out opinions. So, I think it does require some homework at the time of progression but really seek those out.

Lisa Hatfield:

Okay. Thank you. Now, if a patient does have an interest in a clinical trial, say maybe they have, their cancer has progressed, would they seek out that trial through the academic center itself? If, say they live in a rural area and they don’t have access, would they contact the academic center itself, or would they seek out a specialist like you first to ask about those clinical trials?

Dr. Christina Baik:

So they sort of come together in a way, because a lot of the specialists are in academic centers. So I think there are two ways to go about it. One is to meet with the specialist who can give you kind of the landscape of where things are and what might be appropriate. So, that’s one way to do it.  The other way to do it is if there’s a particular clinical trial that you’re really interested in based on discussions with other patients or through advocacy groups, if there are particular clinical trials, usually the contact information is listed on the clinicaltrials.gov website, and the contact number is usually for the research team who can give you more information about that particular trial.

Lisa Hatfield:

Okay. That’s very helpful, thank you. And thank you for this overview. I just want to recap a couple of points that you made that’s really important for patients to know. You had mentioned knowing their type, their subtype of lung cancer, knowing their stage, and knowing their mutations and having an advocate. I think those are all really great tips that you gave.


Share Your Feedback

Create your own user feedback survey

Advanced Prostate Cancer: What You Need to Know About Evolving Treatment and Research

Advanced Prostate Cancer: What You Need to Know About Evolving Treatment and Research from Patient Empowerment Network on Vimeo.

Research is evolving quickly, leading to an increase in treatment options for advanced prostate cancer patients. Expert and researcher Dr. Rana McKay reviews current prostate cancer treatment options, discusses where clinical trials fit into a care plan, and shares advice for partnering with your healthcare team.

Dr. Rana McKay is a Medical Oncologist at UC San Diego Health. Learn more about Dr. McKay.

Download Resource Guide

See More from Evolve Prostate Cancer

Related Resources:

What Factors Impact Head and Neck Cancer Treatment Decisions?

Which Factors Impact Advanced Prostate Cancer Treatment Decisions? 

How to Play an Active Role in Your Prostate Cancer Treatment and Care Decisions

How to Play an Active Role in Your Prostate Cancer Treatment and Care Decisions 

What Prostate Cancer Research Is Showing Promise

How Is Advanced Prostate Cancer Treated? 

Transcript:

Katherine:

Hello and welcome. I’m your host, Katherine Banwell. When advanced prostate cancer patients discussed potential treatment approaches with their healthcare team, it’s important that they understand all of their options including where clinical trials fit in. So, the patient empowerment network created the Evolve Series, to help patients understand the latest research and how it may impact them. In today’s program, we’re joined by a prostate cancer expert who is going to explain and discuss research highlights, and provide tips for having productive conversations about your care. Before we meet our guests, though, let’s review a few important details.  

The reminder email you received about this program contains a link to program materials. If you haven’t already, click that link to access a guide to help you follow along during the webinar. At the end of this program, you’ll receive a link to a program survey. Please take a moment to provide feedback about your experience today in order to help us plan future webinars. And finally, before we get into the discussion, please remember that this program is not a substitute for seeking medical advice.  

Please refer to your healthcare team about what might be best for you. Well, let’s meet our guest today. Joining us, is Dr. Rana McKay. Dr. McKay, welcome. Would you please introduce yourself?  

Dr. McKay:

Absolutely. My name is Rana McKay, I’m a genitourinary medical oncologist, at the University of California in San Diego. It’s a pleasure to be here, with you, on this program today.  

Katherine:

And the sun is shining.  

Dr. McKay:

Yes.  

Katherine:

Which, is always good. Well, thank you so much for taking the time to join us today. Before we get into the discussion, Dr. McKay, I’d like to have you tell us how the landscape of advanced prostate cancer has changed over your career.   

Dr. McKay:

Oh, my goodness. It has absolutely rapidly evolved over the last decade. I think when I was just starting my career, it was right around the time where Abiraterone and Enzalutamide were being heavily tested and just getting approved and entering into the clinic. And as we think about all of the evolution that’s happened since that time, we now have multiple androgen receptor pathway inhibitors in the clinic. We have radioligand therapies in the clinic, radium-223. The first radioligand therapy across any solid tumor malignancy to improve overall survival and on the heels of that, most recently, lutetium PSMA, which is a targeted radioligand therapy.  

There’s several different kinds of chemotherapies, and I think two more diagnostics have evolved. We are now integrating molecular profiling across multiple areas in the disease natural history and actually have several FDA-approved treatments based off of results of molecular profiling, whether that be germline hereditary testing or just tumor testing like, PARP inhibitors and immunotherapy. And then, additionally, to kind of continue on that same thought of our diagnostics changing is one of the greatest disruptors in our treatment of prostate cancer has been the introduction of PSMA PET imaging that has really revolutionized our ability to be able to detect disease at lower levels of PSA.  

And that’s opened up options for focal therapy, radiation therapy, and other sorts of strategies. So, it’s really been just remarkable, all of the different advances that have occurred in prostate cancer over the last decade.  

Katherine:

Well, let’s dive into developing research and what it could mean for patients. Are there recent research highlights that you could share with the audience?  

Dr. McKay:

Absolutely. I think the newest approvals that have occurred in prostate cancer have been the approvals of combination PARP inhibitors, which block the ability of cancer cells to repair their DNA combined with hormonal agents such as abiraterone (Zytiga) or enzalutamide (Xtandi) for patients who have specific mutations in their tumor and their tumor is no longer responding to treatment. Those are the newest FDA approvals that were recently highlighted and shared.  

Katherine:

What areas of research do you specialize in? 

Dr. McKay:

The areas of research that I specialize in are particularly around novel therapeutics for patients with advanced prostate cancer, biomarker development precision medicine strategies for patients across the spectrum. And actually, also, in the localized setting, thinking about how we can attempt to cure more patients with prostate cancer by integrating our systemic therapy with surgical and radiation strategies to improve survival outcomes for patients and ultimately, cure it for patients by using effective systemic therapy early on so patients never recur.   

Katherine:

Yeah. We’ve been hearing a lot recently about innovations in technology. How are these advances accelerating prostate cancer care?  

Dr. McKay:

Innovations of technology have absolutely been revolutionizing prostate cancer care I think from the diagnostic side, there’s new imaging modalities that are getting more refined. On the molecular side, there’s now different kinds of genetic tests. And our ability for us to do these tests, and do them quickly, and get results in real time that we can make decisions on we’ve come a long way from when we first sequenced the human genome. We’re now able to do that so quickly multiple times over in a very streamlined kind of way. And then, I have to say that there’s been tremendous improvement in our modalities of administrating therapies.  

So, our therapies are getting more novel, they’re getting more precise. What I mean, by that is targeted radioligand therapy, targeting linking a small molecule that binds PSMA, labeling it with a payload that is radiation therapy or kind of radio therapy/radio particle doing the same thing with chemotherapy, developing antibody drug conjugates. There’s androgen receptor degraders. There’s different ways of administrating immunotherapy by specific antibodies. So, there’s just the different sorts of treatments that are out there.  

We’ve just come such a long way from hormone therapy, which is still very important in chemotherapy to other different modes of action with the different systemic treatments we have.  

Katherine:

What about individual patients? Is there research into understanding a person’s – just one person’s disease?  

Dr. McKay:

Absolutely. I mean, that is in essence, precision medicine. I think we are now molecularly profiling tumors that is standard of care for anybody with advanced disease to undergo hereditary tumor profiling and – or hereditary profiling of just normal cells in the body, if there’s any sort of genetic abnormalities. But also, the tumor itself, and able to do that all for the actual biopsy specimen, or surgical specimen, and also blood. And then, based off of that individual’s genetic makeup, or the genetic makeup of the tumor, or the immune profile of the tumor actually trying to target therapy.  

There is a clinical trial that we are eagerly developing through the alliance, which we hope will open to enrollment before the end of the year, called the PREDICT Study. And this study is using that very notion of taking somebody’s DNA and RNA from their specific tumor, and based off of their results, strategizing the treatment around what kind of genetic makeup is in the tumor. And I think we’re moving towards that.  

Katherine:

What about common genetic mutations and what are you learning about people who have other genetic mutations like the BRCA mutation?  

Dr. McKay:

For patients who have BRCA mutations, first I’ll say, the prevalence of BRCA mutations varies across the stage of prostate cancer that somebody has. In the localized setting, the prevalence is a lot lower on the order of 2 to 4 percent depending on somebody’s risk profile. In the advanced setting, it is higher, 6 to 8 percent. Patients who have BRCA alterations are particularly susceptible to PARP inhibitors, which are oral drugs that can be given that when given in an individual who’s got a BRCA mutation, can cause cell death; can cause a tumor cell to die. And so, that’s a very good thing.  

 I think the other thing, if thinking about the type of BRCA alterations, if there’s something that’s hereditary, this information is prognostic and predictive in that in can guide how people are going to – how we think they may do and what they may respond to. But it’s also really important because it can inform cascade testing for family members. It could also inform screening for secondary cancers in that individual who has prostate cancer with a known BRCA alteration. So, I think there’s a lot of personalization that happens based off of the molecular profiling results.  

Katherine:

It’s all so exciting, Dr. McKay. But progress can only be made with patient participation in clinical trials, as we know. So, when should a patient consider participating in a trial? 

Dr. McKay:

Thank you so much for bringing this point up. I think our clinical research is critically important to advancing the field. Clinical trials, I think, are really – they offer our patients the treatments of tomorrow today, quite honestly. And I think the way trials are designed, they’re designed to test different treatment modalities, test in reference to the standard of care. I think at any point in time, anybody can think about enrolling on a clinical trial. I think sometimes there’s this false notion that, “I’m not going to enroll in a trial until later on, until I’ve failed all different kinds of treatments.” That’s not true.  

 I think at any juncture along the way where a decision is being made around initiating a systemic therapy, or proceeding with a surgical intervention, or radiation intervention it’s always worthwhile to stop and ask, “Are there any clinical trials that I could be eligible for right now? And if so, what are they? So, I think it’s really important, I think, for patients to know that and to ask of their clinicians that are caring for them, “Are there any clinical trials?”  

And it may be that patients, not to say, may need to travel, but if they’re not necessarily at that institution where somebody may be receiving their care with a clinician asking their doctor, “Are there other trials at places close by where I can go and explore?” I think that’s a really important thing.  

Katherine:

Yeah. That’s good information. What about common misconceptions? What are you hearing from patients about their fears and hesitations about participating in the trial.  

Dr. McKay:

Yeah. I think a lot of patients have a fear of, “I don’t want to be a hamster or a guinea pig. I don’t want to get placebo. I don’t want to get suboptimal care.” So, I think, to step back, I think the clinical trials are designed where actually patients are followed very, very closely, probably even more closely than I think would be in general with laboratory tests, PSA testing, imaging, at critical time points to assess that any therapies or strategies is working. Many trials are not necessarily placebo-controlled trials.  

Placebo-controlled trials are really only utilized in the context when somebody may – where the standard of care is to either do nothing or do one drug alone, not two drugs, and then, somebody’s getting one drug and getting a placebo. So, the placebo-controlled trials are really, first off, they’re later-staged studies, they’re usually Phase III studies, or large Phase II studies that have gotten pretty far on the runway of clinical trial and clinical drug development.  

 And it’s in the context of, you know, “Well, if I didn’t do the clinical trials, I’m probably not going to do anything,” or I’m not going to – you know, “If I decided to not do the trial, I would get no treatment, but if I’m doing the trial, there’s a 50 percent chance I’ll get no treatment and 50 percent chance I may get something. So, we have to think about, “Well, what is the standard of care?” and the standard of care matters because that is what it’s being compared to. If the clinical situation is that the standard of care is to monitor, then that’s where a placebo may be utilized.  

But if a standard of care is that somebody should get treatment with X drug, then that X drug would be in the controlled arm of the study.  

Katherine:

Yeah.  

Dr. McKay:

But not every trial has a placebo.  

Katherine:

What would you say to someone who is nervous or hesitant about participating in a trial?  

Dr. McKay:

Yeah. Very good question, I think. Talk to your clinician. Talk to your doctors about those fears. What are the reservations? What are the concerns? Sometimes, I think the unknown is always – the fear of the unknown kind of causes a lot of angst. But when people are on a clinical trial, when you’re on a clinical trial, you are in control. Some people don’t believe that, but you are, at any point in time, you can decide to stop. You don’t even need to have a reason for why you decide to stop. At any point in time, if something is not working for you, you have choice.   

And so, I think that is something that is really important for patients to know that you’re actually in control, you’re being watched very closely, being watched very carefully for safety toxicity. If there’s a toxicity, people are not going to – you’re not going to just stay getting the same regiment in the exact same way if you’re not tolerating it. If something isn’t working, you’re not going to continue receiving the therapy that’s no longer working just because you’re on a clinical trial. 

Katherine:

Right.  

Dr. McKay:

And you’re in control; at any point in time, you could say, “I don’t want to participate anymore.”  

Katherine:

Yeah. Are there barriers for accessing trials? And if so, do you have any recommendations for how to tackle those?  

Dr. McKay:

Yeah. I think there are barriers to accessing trials. I think it can be very overwhelming because there’s thousands of clinical trials that are being conducted for people with prostate cancer. And I think as a patient, sometimes it’s hard to navigate that. But I think the thing to take home is that you do not have to do it alone, and you should not do it alone because I think half of the trials that are out there, the large bulk of them may not necessarily be directly applicable to you or relevant for you.  

 And so, I think talking to your clinician about that, I think seeking care, even if just for a second opinion at an NCI-designated cancer center, or NCI-designated comprehensive cancer center is probably a good idea. You know, if you’re hearing the same message from your local clinician then that’s great. If there’s more options that are being presented to you, that’s great, those are more options that you could tap into. I think talking to patients who have gone on a trial may also help away some of the fear around participating in a clinical trial, and there’s lots of platforms where that could take place either asking your physician, or the American Cancer Society, or other societies can help connect patients to one another.  

Katherine:

Okay. I’m glad you mentioned some of the resources because that’s what I was going to ask you about. Well, I want to mention to our audience that if you want more basic information about prostate cancer, PEN has created a prostate cancer toolkit, which includes information about diagnosis and staging. And you can find it at powerfulpatients.org.  So, before we move onto understanding current treatment options, Dr. McKay, what are the goals of advanced prostate cancer treatment? And how do they vary by patient?  

Dr. McKay:

Yeah. I do think the goals can vary. I think in my mind, a lot of times, it’s making people live longer, making them feel better. Those are the two salient goals and if our therapies are not achieving one or other of those two goals then we need to rethink the strategy. But different people are different, and they may weigh the risks and benefits of any given therapy, or the slated benefit with the slated risk through a different lens. And I think it’s critically important to ensure that you’re having those communications with your doctor about the things that matter to you and the things that are really important to you. 

Especially, for people who have advanced prostate cancer. So, I think that can help your clinician strategize, “Okay, is this an individual who wants the kitchen sink everything that I can do even if that means more toxicity that I’m going to offer this thing? Or is this a situation where, you know what, unless there’s data that the kitchen sink is going to work, I really kind of want to temper things and try an approach that’s going to be effective, but maybe not associated with that degree of toxicity.” So, those kinds of conversations absolutely need to be happening.   

Katherine:

Yeah. With all the recent advances in treatment, is there a standard approach now to treating someone with advanced disease? And if so, what is it?  

Dr. McKay:

Yeah. There absolutely is a standard approach. There’s guidelines that are based off of the FDA-approved regimens of the different agents that can be utilized. There’s data regarding sequencing though, I think there’s more data that needs to be had on sequencing. There are guidelines on when to do germline testing, when to do tumor profiling, when to integrate PSMA PET imaging, the standard hormonal agents, who to use them. So, I do think that there are – there’s a set framework of appropriate management and treatment. But there’s a lot of personalization that is overlaid on top of that rubric. And I think that’s the art of medicine.  

Katherine:

Right. Is there testing to understand if a patient’s disease is more aggressive? Or maybe will respond to a certain type of therapy before you begin it?  

Dr. McKay:

Yeah. A very good question. And I think predictive biomarkers, as you described them, there are several for men with prostate cancer, but there’s not a ton of them. So, we know that homologous recombination repair alterations, HRR, gene alterations, particularly BRCA 1, 2, probably 2, we know that those are biomarkers of response to PARP inhibitors. We know that patients who have high tumor mutation burden, or have a mismatch repair, that those are markers of response to immunotherapy. We know that if people have a certain level of PSMA PET vividity on their PET scan, that that’s a biomarker for receiving lutetium PSMA.  

Those are the main biomarkers that are actually in use in the clinic to date. But I think there’s a lot more that I think are being explored from mutations in the androgen receptor, or amplifications in the androgen receptor, being potentially predictors of response to different degraders, different kind of hormonal agents. There’s certain tumor suppressor gene mutations that may predict that patients may do a little bit better with chemotherapy. So, there’s other markers that are being looked at, but they don’t have the same robustness as the BRCA 1, 2 and other ones that I talked about. 

Katherine:

Yeah. How does a patient’s health and lifestyle impact what treatment approach is right for them?  

Dr. McKay:

I mean, health and lifestyle, diet, and exercise, nutrition, sleep are so important. I think that one of the backbones of treatment for hormonal therapy is androgen deprivation therapy. There can be negative consequences with regards to muscle mass, bone mass, other things related to that therapy. So, I think it’s critically important for patients to maintain a healthy diet, making sure they’re getting appropriate exercise, weight-bearing, resistance training.  

And I think, too, this helps people with their functionality, with their ability, their reserve, and ability to tolerate treatment or tolerate more aggressive treatment. So, half of my clinic is talking about diet and exercise, and how to optimize individual health when people are on therapy. 

Katherine:

Yeah. Mentally, a good diet and sleep –  

Dr. McKay:

Yes.  

Katherine:

And exercise is going to be helpful.  

Dr. McKay:

Yes.  

Katherine:

As well. What about comorbidities? Do they play a role?  

Dr. McKay:

They absolutely do play a role. I think comorbidities like cardiovascular disease, diabetes absolutely can play a role. The hormone therapies, patients can have a propensity to gain weight, they can have a propensity to have worsened cholesterol being on hormone therapy, which can then affect somebody’s cardiovascular health. And so, some of the drugs cause increased hypertension. So, I think understanding the different comorbidities that any individual may have is important in selecting the best therapy, “Well, actually, if you’ve got X, Y, Z going on, maybe I’m going to shy away from this, but lean more towards that.”  

I think making sure that your physician knows about that and knows about changes that happen along the way. Sometimes, people with prostate cancer, many a times they have a long, natural history where they’re seeing the physician caring for them for their prostate cancer over many, many years. And somebody’s medical history, when they first saw that individual, it’s going to change and evolve over time as different things happen. And so, I think keeping your clinician that’s caring for you for your prostate cancer informed of all the other non-cancer things that are happening I think is a really good idea.  

 If you had a fracture, that’s actually a really important thing for somebody who’s got prostate cancer. Or “Gosh, my primary care just started me on Metformin because they think my blood sugar is a little bit off.” These are important things, I think, for clinicians to know about.  

Katherine:

Yeah. It’s all about communication, isn’t it?  

Dr. McKay:

Absolutely. Yeah.  

Katherine:

Don’t worry about over-sharing.  

Dr. McKay:

Yeah.  

Katherine:

Yeah. Speaking of sharing, shared decision-making has become the gold standard, really, for encouraging a successful relationship between a patient and their healthcare team. What does shared decision mean to you as a provider?  

Dr. McKay:

Yeah. I think shared decision is an open dialogue. I think it’s an open dialogue with the physician, with the patient, sometimes, often times, the patient’s caregivers, and families, and loved ones may be involved in that process, where we’re talking about, first off, establishing the goals. Well, what are the goals? And I think, when we start with the goals then, we can say, “Okay. Well, what are the things that we can do to achieve those goals?” And I think sometimes we just dive right into, “Well, what are we going to do with the next step?”  

So, I think establishing what the goals of therapy are the things that matter to any individual patient and their family is important. And then, from there, working on, “Okay. Well, aligning with those goals, these are the different things that you can do. These are the pros and cons of the different things that you could do,” and making an informed decision about the next step.  

Katherine:

What questions should a patient ask about potential treatment options?  

Dr. McKay:

One, what are the different treatment options? You know, sometimes I think that statement doesn’t get said enough. What are the standard of care options? What are the clinical trial options? Ask are there radiation therapies, surgical options? That may be a relevant question for some individuals, some individuals, not. Being very open like, “Okay, I’m hesitant about chemo. Let me explore that.” Well, where does that hesitancy stem from? What’s the fear about chemo? Are there chemotherapy-sparing options right now? Or how can we kind of dispel the fear or myth around chemotherapy?  

So, I think these are the questions that I think a patient can ask. How is a therapy administered? Where do I go? How would I receive different therapies are given at different modes of administration? I think those are good questions. Who do I call if something happens to me on the weekend or on a holiday? Who do I reach out to? What are the phone numbers? Give me all the phone numbers. Get them in my phone. Save them in there, so you know, who to reach out to if you ever need something, if you ever need assistance.  

Katherine:

Yeah, that’s really good advice. Why should a patient consider finding a prostate cancer specialist?  

Dr. McKay:

I think a patient should consider finding a prostate cancer specialist because quite honestly, the field of oncology is getting to be so expansive, and there’s so many changes in guidelines on a monthly basis, sometimes across all the different malignancies. So, I think having a specialist who understands the nuances of the different iterations of treatment for people with prostate cancer, and how to personalize that for a given patient is really important. And I think it can be associated with improved outcome.  

I will say that the note about clinical trials, there have been several studies that had been conducted that have actually noted that patients who enroll on a clinical trial, whether or not that clinical trial is positive or not, independent of the results of the trial. But just enrolling on a clinical trial is associated with improved outcome. And I think a lot of it stems with where people get their care, eligibility for trials, the scrutiny that happens when people are on trials, and sort of, level of expertise where people get their care and so forth.  

Katherine:

Yeah. Thank you for sharing all of this information, Dr. McKay, it’s really vital. As we close, what final thoughts would you like to leave our audience with? Why are you hopeful?  

Dr. McKay:

I am very hopeful because of all of the amazing technologies that are in the pipe right now, currently in development, some early on, some close to the finish line that I think are certainly going to change the way that we view and treat prostate cancer. I think it’s exciting to see where the field has come and where the field is going, and know that you are not in this alone, and there’s a lot of progress that is being made, and a lot of hope that is out there for individuals who have prostate cancer.  

Katherine:

Well, Dr. McKay, thank you so much for taking the time to join us today. We really appreciate it.  

Dr. McKay:

Wonderful. It’s my pleasure.  

Katherine:

And thank you to all of our collaborators.  

If you’d like to watch this webinar again, there will be a replay available soon. You’ll receive an email when it’s ready. And don’t forget to take the survey immediately following the webinar. It will help us as we plan future programs. To learn more about prostate cancer and to access tools to help you become a proactive patient, visit powerfulpatients.org. I’m Katherine Banwell. Thanks for being with us.  

Lung Cancer Biomarker Disparities | How Precision Medicine and Research Can Help

Lung Cancer Biomarker Disparities | How Precision Medicine and Research Can Help from Patient Empowerment Network on Vimeo.

How can lung cancer research and precision medicine help with biomarker disparities? Experts Dr. Joshua Sabari from NYU Langone and Dr. Eugene Manley from SCHEQ Foundation discuss testing factors that need improvement, patient groups that show disparities, and how clinical trial participation can move research forward.

[ACT]IVATION TIP

“…we really have to more universally test everyone equally to really have an impact on outcomes.”

Download Resource Guide  | Descargar guía de recursos

See More from [ACT]IVATED NSCLC Biomarkers

Related Resources:

Hope Unleashed: Advancing Therapies for Defiant Mutations in Lung Cancer

Hope Unleashed: Advancing Therapies for Defiant Mutations in Lung Cancer

Maximizing Biomarker Equity: Leveraging Partnerships to Close Biomarker Disparities in Lung Cancer

Maximizing Biomarker Equity: Leveraging Partnerships to Close Biomarker Disparities in Lung Cancer

How Can We Leverage Lung Cancer Biomarker Data to Address Health Disparities?

How Can We Leverage Lung Cancer Biomarker Data to Address Health Disparities

Transcript:

Lisa Hatifeld:

So, how can advancements in precision medicine be made more inclusive and equitable to ensure that biomarker-driven treatments benefit diverse populations equally? Second part is, what do you see as the most pressing research priorities in understanding and mitigating these biomarker disparities?

Dr. Joshua Sabari:

So I think first and foremost, testing is key. I mean, educating clinicians, healthcare providers, that every single patient, no matter what clinical characteristic that may be, age, sex, ethnicity, race needs to be tested broadly with the same mutational sort of profile or same biomarker profile. Having somebody in your office who never smoked, those patients generally will have broad panel and next generation sequencing. If you have an 85-year-old patient who is a former heavy smoker, the rate of mutational testing comes down.

So I think we need to remove that bias, that those clinical biases that we have, that we carry with us on a day-to-day basis. We need to test all patients with lung cancer regardless of any clinical characteristic. And what I tell my fellows, my residents, and what I talk to patients about is really all you need is lungs to develop lung cancer.

We need to remove that stigma and when we remove that stigma, we will be testing more broadly in our practices. There are also a lot of systemic biases, a lot of racism that exists, that prevents clinicians, I believe, from doing the best thing for their patients. And if you look at clinical trial enrollment in this country and that’s something that we do need to improve in order to develop better treatment options for our patients, particularly our patients of Latin American descent or Black Americans in the United States.

We need to enroll more patients of more diverse backgrounds onto our trials. Otherwise, we’re only limiting our treatments to specific or small percent of our patient population. So to be honest, I don’t know how well our EGFR inhibitors work in Black patients. I know it’s approved and we utilize it, but we don’t have nearly as much data prospectively treating novel therapies.

A lot of our trials have inclusion rates as low as 2 percent to 3 percent. And we know that our Black patient populations make up 13 percent to 15 percent of our practices. So I think more needs to be done to align our enrollment on trial, I think from institutional policies as well as governmental. So the FDA has really made a forceful statement here to pharmaceutical companies that if your data is not inclusive of a U.S. patient population, this will have ramifications for approvals in the future.

So a lot needs to be done in the sense of education both from the healthcare provider and…but also from the patient, and to really motivate patients to enroll in trials. And one positive that I’ve seen from the patient support groups, the advocacy groups, particularly EGFR Resisters Group, for example, we’ve seen a tremendous sort of push for patients to enroll on trials, again, to benefit themselves as an individual patient diagnosed with EGFR mutant lung cancer, but also to help those who come before or after them in their journey with lung cancer.

Lisa Hatfield:

And, Dr. Manley, do you have anything to add to that?

Dr. Eugene Manley:

I think he hit most of it, but I will say that you have to test everyone because there are people that have risk factors for lung cancer and those that don’t. And like, one of the leading risk factors is history of smoking, but there’s a significant population of specifically Asian females that don’t smoke. Even recently, that have been showing that Black women that don’t smoke also have increased rates of lung cancer. And these are, we don’t know why.

So we still need to be able to test all these patients across all the indications and maybe cross-reference with stress income, socioeconomic status and really try to determine maybe if there are certain specific drivers and what we didn’t talk about. We know that there are some epigenetic changes that may occur due to stress. We also know that there are some changes in tumor mutational burden, some stuff out of MSK. And I think there is some stuff that even shows differences in the immunomarker frequency and response in Black populations. So, we really have to more universally test everyone equally to really have an impact on outcomes.


Share Your Feedback

Create your own user feedback survey

Equity in Action | Addressing Biomarker Disparities in Lung Cancer

Equity in Action: Addressing Biomarker Disparities in Lung Cancer from Patient Empowerment Network on Vimeo.

How can biomarker disparities be reduced in lung cancer patients? Experts Dr. Joshua Sabari from NYU Langone and Dr. Eugene Manley from SCHEQ Foundation discuss approaches that are being used for community engagement and further interventions that can be used to reduce disparities.

Download Resource Guide  | Descargar guía de recursos

See More from [ACT]IVATED NSCLC Biomarkers

Related Resources:

Understanding Non-Small Cell Lung Cancer: Types, Biomarkers, and Treatment Insights

Understanding Non-Small Cell Lung Cancer: Types, Biomarkers, and Treatment Insights

Navigating Lung Cancer Biomarker Testing | Challenges and Solutions for Timely Access

Navigating Lung Cancer Biomarker Testing | Challenges and Solutions for Timely Access

When Should Lung Cancer Patients Receive Biomarker Testing?

When Should Lung Cancer Patients Receive Biomarker Testing?

Transcript:

Lisa Hatfield:

So, Dr. Manley, are there any promising approaches or interventions aimed at reducing biomarker disparities that you’ve currently been exploring or are advocating for?

Dr. Eugene Manley:

I will take several angles on this. One thing is there has to be much more community engagement and involvement and really going to community groups, whether they’re faith-based, whether they’re barbershops, really going out where people are and letting them know about lung cancer, lung cancer disparities, biomarker testing, what you can do. The other way is also going to conferences where there are more diverse scholars that are attending. So a lot of these are STEMM meetings. They may not be specific in lung cancer, but if you can go out there and get the word out about lung cancer and the disparities, then they can go back to their families and talk about, you know, screening and testing and making sure that their family members are aware.

And then, you know, we just published a paper recently that shows the upstream part of biomarker testing is where are we starting at with our cell line? We just did a review of all the lung cancer cell lines. Of over 800 cell lines, majority were European-based. Only 31 cell lines in total were from Black African American populations. None were from Hispanic, none were from Native American, Pacific Islander, none from Alaska Native.

So just think about this. If that is our starting material for all of our biomarker testing and TCGA and databases, then everything we’re developing is on a population that already has great access and outcomes. But they don’t have the greatest disparities. So then you’re getting through doing all these trials, and then you have biomarkers, and you have immunotherapies coming out, and then you’re seeing adverse events in these diverse populations at the end because you don’t have the starting material.

Lisa Hatfield:

And, Dr. Sabari, after hearing Dr. Manley’s comments about that, how do you…or do you know of any approaches or interventions that are aimed at reducing these biomarker disparities? Because maybe they aren’t being acknowledged yet. Maybe they’re only being seen in certain populations.

Dr. Joshua Sabari:

Yeah, I think Dr. Manley hit it on the head. First off, we don’t even know the correct or true numbers for certain mutations in specific patient populations. And I just read an article about patients from Latin America, different rates of EGFR, ALK, and other mutations. You can imagine a study population from Africa, for example. And then obviously studying a population of Black Americans here in the United States as well.

We know that most of the cell lines, most of the data that we’ve had, particularly TCGA  (Tumor Cell Genome Atlas) is from a Caucasian or North European patient population. So I think we need to do better in that sense. I think equally as important, are clinical trial enrollment needs to diversify. Again, it’s mostly women. It’s mostly Caucasian women. We have very, very low rates of Hispanic patients enrolled on clinical trials, Blacks enrolled on clinical trials.

So I think we need to do better in that sense. One thing that we’ve really pushed for in academic medicine is to at least report who is being enrolled on trials so that we can understand is this data generalizable to my own clinical practice? And oftentimes if you look at the clinical characteristics of patients enrolled on the trial, it likely does not match what you see in your own practices.

So we need to do better in that sense. So I think the FDA, and especially pharmaceutical companies, are clearly looking to expand and broaden their inclusion criteria and also access to patients so that we can actually have a more diverse patient population that represents our country enrolled on these trials.


Share Your Feedback

Create your own user feedback survey